MXPA02007058A - Indazole compounds, pharmaceutical compositions, and methods for mediating or inhibiting cell proliferation. - Google Patents

Indazole compounds, pharmaceutical compositions, and methods for mediating or inhibiting cell proliferation.

Info

Publication number
MXPA02007058A
MXPA02007058A MXPA02007058A MXPA02007058A MXPA02007058A MX PA02007058 A MXPA02007058 A MX PA02007058A MX PA02007058 A MXPA02007058 A MX PA02007058A MX PA02007058 A MXPA02007058 A MX PA02007058A MX PA02007058 A MXPA02007058 A MX PA02007058A
Authority
MX
Mexico
Prior art keywords
mmol
compound
nmr
pharmaceutically acceptable
prodrug
Prior art date
Application number
MXPA02007058A
Other languages
Spanish (es)
Inventor
Siegfried Heinz Reich
Original Assignee
Agouron Pharma
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Agouron Pharma filed Critical Agouron Pharma
Publication of MXPA02007058A publication Critical patent/MXPA02007058A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/081,2,4-Triazoles; Hydrogenated 1,2,4-triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B22CASTING; POWDER METALLURGY
    • B22DCASTING OF METALS; CASTING OF OTHER SUBSTANCES BY THE SAME PROCESSES OR DEVICES
    • B22D41/00Casting melt-holding vessels, e.g. ladles, tundishes, cups or the like
    • B22D41/50Pouring-nozzles
    • B22D41/56Means for supporting, manipulating or changing a pouring-nozzle
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/12Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/54Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings condensed with carbocyclic rings or ring systems
    • C07D231/56Benzopyrazoles; Hydrogenated benzopyrazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/56Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic Table
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/18Compounds having one or more C—Si linkages as well as one or more C—O—Si linkages
    • C07F7/1804Compounds having Si-O-C linkages

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Mechanical Engineering (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

Indazole compounds that modulate andor inhibit cell proliferation, such as the activity of protein kinases are described. These compounds and pharmaceutical compositions containing them are capable of mediating , e.g., kinasesdependent diseases to modulate andor inhibit unwanted cell proliferation. The invention is also directed to the therapeutic or prophylactic use of pharmaceutical compositions containing such compounds, and to methods of treating cancer as well as other disease states associated with unwanted angiogenesis andor cellular proliferation, such as diabetic retinopathy, neovascular glaucoma, rheumatoid arthritis, and psoriasis, by administering effective amounts of such compounds.

Description

INDA ^ OL COMPOUNDS, PHARMACEUTICAL COMPOSITIONS AND METHODS FOR THE MEDIATION OR INHIBITION OF CELL PROLIFERATION Field of the invention This invention is directed to indazole compounds that mediate or inhibit cell proliferation, for example, through the inhibition of the activity of protein kinases, such as VEGF, CHK-1, and cyclin-dependent kinases (CDK). , such as CDK1, CDK2, CDK4, and CDKß. The invention extends to pharmaceutical compounds containing such compounds and compositions, and to cancer treatment methods as well as other pathological conditions associated with undesired angiogenesis and / or cell proliferation, by administration of sufficient amounts of such compounds.
BACKGROUND OF THE INVENTION Uncontrolled cell proliferation is the hallmark of cancer. Cell proliferation in response to various stimuli is manifested by the deregulation of the cell division cycle, the process Ref: 138171 by which the cells multiply and divide. Tumor cells typically have a lesion of genes that directly or indirectly regulate progression throughout the cell division cycle.
Hyperproliferative disease states, including cancer, are characterized by unbridled circulation through the cell cycle with uncontrolled vigor due to, for example, injury to genes that directly or indirectly regulate progression through the cycle. Therefore, agents that modulate the cell cycle, and therefore hyperproliferation, could be used to treat various pathological conditions associated with uncontrolled or unwanted cell proliferation. In addition to cancer chemotherapeutic agents, cell cycle inhibitors are postulated as antiparasitic (See, Gray et al., Curr. Med. Chem. 6, 859-875 (1999)) and have recently been demonstrated as antiviral potentials (See , Schang et al., J. Virol. 74, 2107-2120 (2000)). Moreover, the utility of antiproliferative agents can be extended to the treatment of cardiovascular disorders such as atherosclerosis or restenosis (See Braun-Dullaeus et al., Circul a ti on, 98, 82-89 (1998)), and inflammatories such as arthritis (See, Taniguchi et al., Na ture Med., 5, 760-767 (1999)) or psoriasis.
The mechanisms of cell proliferation are being actively investigated at the cellular and molecular level. At the cellular level, deregulation of the pathways, loss of control of the cell cycle, unrestrained angiogenesis or stimulation of inflammatory pathways are under scrutiny, while at the molecular level, these processes are modulated by various proteins, among which the Protein kinases are prominently suspected. The overall decrease in proliferation can also be a consequence of programmed cell death, or apoptosis, which is also regulated by multiple pathways, some of which involve proteolytic enzyme proteins.
Among the regulatory proteins postulated, protein kinases are a family of enzymes that catalyze the phosphorylation of the hydroxyl group of specific tyrosine, serine or threonine residues in proteins. Typically, said phosphorylation drastically disrupts protein function, and therefore protein kinases are the main axis of regulation of a wide variety of cellular processes, including metabolism, cell proliferation, differentiation cellular, and cell survival. Of the numerous different cellular functions in which it is known that the activity of protein-qumasa is necessary, some processes represent attractive targets for therapeutic intervention for certain pathological states. Two examples are cell cycle control and angiogenesis, in which protein kinases play a central role; Both processes are essential for the growth of solid tumors and also in other diseases.
CDKs constitute a class of enzymes with a critical role in regulating the transition between different phases of the cell cycle, such as the progression of a quiescent Gi state (the lapse between mitosis and the start of DNA replication for a new beginning of cell division) to the S state (the period of active synthesis of DNA), or the progression of the G2 phase to the M phase, in which active mitosis and cell division occur. See, for example, the articles compiled in Sci ence, vol. 274 (1996), pp. 1643-1677; and Ann. Rev. Cel l Dev. Bi ol. , vol. 13 (1997), pp. 261-291. CDK complexes are formed by the association of a regulatory cyclin subunit (eg, cichna A, Bl, B2, DI, D2, D3, and E) and a catalytic qumase subunit (eg, cdc2 (CDK1), CDK2, CDK4, CDK5 and CDK6). As the name implies, CDKs have a total dependence on the cyclin subunit to phosphorylate their specific substrates, and different pairs of kinases and cyclins function in the regulation of progression through the different phases of the cell cycle.
Cyclins D are sensitive to extracellular growth signals and become activated in response to mitogens during the Gi phase of the cell cycle. CDK4 / cyclin D plays an important role in the progression of the cell cycle by phosphorylating, and therefore inactivating, the retinoblastoma (Rb) protein. The hypophosphorylated Rb binds to a family of transcriptional regulators, but as Rb is hyperphosphorylated by CDK4 / cyclin D, these transcription factors are released and activate genes whose products are responsible for the progression of the S phase. Phosphorylation of the Rb and the inactivation by CDK4 / cyclin D allows the passage of the cell beyond the restriction point of the Gi phase, after which the sensitivity to extracellular growth or inhibitory signals is lost, and the cell is dedicated only to the division. During the late Gi phase, Rb is also phosphorylated and inactivated by CDK2 / cyclin E, and recent evidence indicates that CDK2 / cyclin E can also regulate the progression towards the S phase through a parallel pathway that is independent of the phosphorylation of Rb (see Lukas et al., "Cyclin E-induced S Phase Without Activation of the pRb / E2F Pathway", Genes and Dev. , Vol 11 (1997), pp. 1479-1492).
The progression of the Gi phase to the S phase, achieved by the action of CDK4 / cyclin D and CDK2 / cyclin E, is subject to a variety of growth regulating mechanisms, both negative and 10 positives Stimuli for growth, such as mitogens, cause an increase in cyclin DI synthesis and thus an increase in functioning CDK4. In contrast, cell growth can be "restrained" in response to DNA damage or negative stimuli 15 for growth, by induction of endogenous inhibitory proteins. These natural inhibitory proteins include the families p21WAF 1 / CIP1, P27KIP and pl6INK4, the latter of which exclusively inhibits CDK4 (see Harper, "Cyclin Dependent Kinase Inhibitors", 20 Cancer Surv. , vol. 29 (1997), pp.91-107). The aberrations in this control system, particularly those that affect the function of CDK4 and CDK2, are involved in the advance of the cells towards the highly proliferative state characteristic of the cells. 25 neoplasms, such as familial melanomas, carcinomas "SBÉSSEBS: X. of esophagus, and pancreatic cancer (see, for example, Hall and Peters, "Genetic Alterations of Cyclins, Cyclin-Dependent Kinases, and CDK Inhibitors in Human Cancer," Adv. Cancer Res., vol 68 (1996), pp.67-108; and Ka by col., "A Cell Cycle Regulator Potentially Involved in Genesis of Many Tumor Types," Sci ence, vol 264 (1994), pp. 436-440). Overexpression of cyclin DI is linked to esophageal, breast and epidermoid carcinomas (see, for example, DelSal et al., "Cell Cycle and Cancer: Critical Events at the Gi Restriction Point", Cri ti cal Rev. Oncogenesi s, vol. 71 (1996), pp. 127-142). The genes encoding the specific inhibitors of CDK4 of the plß family frequently have deletions and mutations in familial melanoma, gliomas, leukemias, sarcomas, pancreatic carcinoma, non-small cell lung carcinoma, and carcinomas of the head and neck (see Nobori et al., "Deletions of the Cyclin-Dependent Kinase-4 Inhibitor Gene in Multiple Human Cancers," Na ture, vol 368 (1994), pp. 753-756). The amplification and / or overexpression of cyclin E has also been observed in a wide variety of solid tumors, and high levels of cyclin E have been correlated with unfavorable prognoses. In addition, cellular levels of the p27 inhibitor of CDK, which acts as a substrate and inhibitor of CDK2 / cyclin E, are abnormally low in breast, colon and prostate carcinomas, and the expression levels of p27 correlate inversely with the stage of the disease (see Loda et al., "Increased Protease Dependent Degradation of the Cyclin-Dependent Kinase Inhibitor p27 in Aggressive Colorectal Carcinomas, "Na t ure Medi cine, vol 3 (1997), pp. 231-234). There is recent evidence that CDK4 / cyclin D could abduct p27, as reviewed by Sherr, et al., Genes Dev., Vol. 13 (1999), p. 1501-1512. The p21 proteins also appear to transmit the tumor suppression signal p53 to the CDK; thus, mutations in p53 in approximately 50% of all human cancers can indirectly result in deregulation of CDK activity.
Emerging reports provide strong validation for the use of compounds that inhibit CDK, and CDK4 and CDK2 in particular, as anti-proliferative therapeutics. Certain biomolecules have been proposed for this purpose. For example, U.S. Pat. No. 5,621,082 to Xiong et al. discloses the nucleic acid coding of the CDK6 inhibitors, and WO 99/06540 of the CDKs. Peptides and peptidomimetic inhibitors are described in European Patent Publication No. 0 666 270 A2, Bandara, et al., Na t uie Bi otechnol ogy, Vol. 15 (1997), pp. 896-901 and Chen, et al., Proceedings of the Na tional Academy of Sci ence, USA, Vol. 96 (1999), p. 4325-4329. Peptide aptagen were identified by screening in Cohen et al., Proc. Na ti. Acad. Sci. U. S TO . , Vol. 95 (1998), pp.14272-14277. Several small molecules have been identified as CDK inhibitors (for recent reviews, see Webster, "The Terapeutic Potential of Targeting the Cell Cycle," Exp. Opin., Investigations, Vol. 7 (1998), pp. 865-887 , and Stover, et al., "Recent advances in protein kinase inhibition: current molecular scaffolds used for inhibitor synthesis", Curren t Opinion in Drug Di verse and Devel opmen t, Vol.2 (1999), pp. 274-285 ). The flavopiridol exerts modest selectivity for the inhibition of CDKs and other kinases, but inhibits CDK4, CDK2 and CDK1 equipotentially, with IC5o ranging from 0.1-0.3 μM. Flavopiridol is currently in Phase II clinical trials as a cancer chemotherapeutic (Sedlacek et al., "Flavopiridol (L86-8275; NSC 649890), A New Kinase Inhibitor for Tumor Therapy", Int. J. Oncol., Vol.9 (1996), pp. 1143-1168). Flavopiridol analogs are subject to other publications, for example, U.S. Pat. No. 5,733,920 of Mansuri et al. (International Publication No. WO 97/16447) e International Publication No. WO 97/42949 and WO 98/17662. The results with purine base derivatives are described in Schow et al., Bioorg. Med. Chem. Lett. , Vol. 7 (1997), pp. 2697-2702; Grant et al., Proc. Amer. Assoc. Cancer. Res., Vol. 39 (1998), Abst. 120 ?; Legravend et al., Bioorg. Med. Chem. Lett., Vol.8 (1998), pp. 793-798; Gray et al., Science, Vol. 281 (1998), pp. 533-538; Chang, et al., Chemistry & Biology, Vol. 6 (1999), pp. 361-375, WO 99/02162, WO 99/43675 and WO 99/43676. Additionally, the following publications disclose certain pyrimidines that inhibit cyclin-dependent kinases and kinases mediated by growth factors: International Publication No. WO 98/33798; Ruetz et al., Proc. Amer. Assoc. Cancer Res., Vol. 39 (1998), Abst. 3796; and Meyer et al., Proc, Amer. Assoc. Cancer. Res., Vol. 39 (1998), Abst. 3794 Benzenesulfonamides which block cells in Gl phase are under development by Eisai, see Owa, et al., J. Med. Chem., Vol. 42 (1999), pp. 3789-3799. An oxindolic inhibitor of CDKs is under development by Glaxo-Wellcome, see Luzzio, et al., Proc. Amer. Assoc. Cancer Res., Vol. (1999), Abst. 4102 and WO99 / 15500. The paullones were found in collaboration with the NCI, Schultz, et al., J. Med. Chem., Vol. (1999), pp. 2909-2919. The indenopyrazones are described in W099 / 17769 and by Seltz, et al, 218. tLha ACS Na ti. Mtg. (August 22-26, 1999, New Orleans), Abst MEDI 316. Aminothiazoles are used in W099 / 24416 and W099 / 21845.
CHK1 is another protein kinase. CHK1 plays an important role as a checkpoint in the progression of the cell cycle. These control points coordinate the progression of the cell cycle by influencing the formation, activation and subsequent inactivation of the cyclin-dependent kinases. The control points prevent the progression of the cell cycle at inappropriate times, maintain the metabolic balance of the cells while they are stopped and, in some cases, can induce apoptosis (programmed cell death) when the requirements of the control point are not met. See, for example, O'Connor, Cancer Surveys, 29, 151-182 (1997); Nurse, Cell, 91, 865-867 (1997); Hartwell et al., Sci ence, 266, 1821-1828 (1994); Hartwell et al., Sci ence, 246, 629-634 (1989).
A series of control points monitor the integrity of the genome and, by sensing DNA damage, these "DNA damage control points" block the progression of the cell cycle in the Gi and G2 phases, and slow down the progression through the phase S.
O'Connor, Cancer Surveys, 29, 151-182 (1997); Hartwell et al., Sci ence, 266, 1821-1828 (1994). This action allows DNA repair processes to complete their tasks before genome replication occurs and the subsequent separation of their genetic material into new daughter cells. Importantly, the gene most commonly mutated in human cancer, the p53 tumor suppressor gene, produces a DNA damage control protein that blocks the progression of the cell cycle in the Gi phase and / or induces apoptosis (programmed cell death) after DNA damage. Hartwell et al., Sci ence, 266, 1821-1828 (1994). It has also been shown that the p53 tumor suppressor strengthens the action of a DNA damage checkpoint in the G2 phase of the cell cycle. See, for example, Bunz et al., Sci ence, 28, 1497-1501 (1998); Wmters et al., Oncogene, 17, 673-684 (1998); Thompson, Oncogene, 15, 3025-3035 (1997).
Given the importance of p53 as the natural axis of the tumor suppression pathway in human cancer, therapeutic interventions that exploit vulnerabilities in cancer with defective p53 have been intensively sought. An emerging vulnerability lies in the operation of the G2 control point in neoplastic cells with defective p53. The cancer cells, »4? F * lacking control point Gi, they are particularly vulnerable to the lack of the last remaining barrier of protection against the effects of DNA damage caused by cancer: the G2 checkpoint. The control point G2 is regulated by a control system that has been preserved from yeasts to humans. In this conserved system, a chemase, CHK1, is important, which transduces signals from the DNA damage sensor complex to inhibit the activation of cyclin B / Cdc2 kinase, which promotes entry into the mitosis phase. See, for example, Peng et al., Sci ence, 277, 1501-1505 (1997); Sánchez et al., Sci ence, 277, 1497-1501 (1997). Inactivation of CHK1 has been shown to abrogate G2 arrest induced by DNA damage both endogenous and caused by both anticancer agents, as well as result in preferential death of the resulting defective cells at the control point. See, for example., Nurse, Cell, 91, 865-867 (1997); Welnert, Sci ence, 277, 1450-1451 (1997); Walworth et al., Na t ure, 363, 368-371 (1993); and Al-Khodairy et al., Mol eq. Bi ol. . Cell, 5, 147-160 (1994).
Selective manipulation of the checkpoint in cancer cells could provide extensive use of chemotherapeutic and antieoplastic radiotherapeutic regimens and could, in addition, provide a common hallmark of the "genomic instability" of human cancer to be exploited as a selective basis for the destruction of cancer cells. A number of factors place CHKl as the central target at the point of control of DNA damage. Elucidation of inhibitors of this and other functionally related kinases such as CDS1 / CHK2, a kinase that was recently discovered to be a cooperator with CHK1 in the regulation of S-phase progression (see Zeng et al., Na t ure, 395, 507-510 (1998); Matsuoka, Sci ence, 282, 1893-1897 (1998)), could lead to new therapeutic modalities in the treatment of cancer.
Another group of chemists are tyrosine kinases. The tyrosmo-kinases can be of the receptor type (having extracellular, transmembrane and intracellular domains) or of the non-receptor type (being totally intracellular). At least one of the non-receptor protein tyrosmaqumases, LCK, is supposed to mediate the transduction in T cells of a cross-interaction signal between a cell surface protein (Cd4) and an anti-Cd4 antibody. A more detailed discussion of non-receptor tyrosine kinases is provided by Bolen, Oncogene, 8, 2025-2031 (1993), which is incorporated herein by reference.
In addition to their role in controlling the cell cycle, protein kinases play a crucial role in angiogenesis, which is the mechanism by which new capillaries are formed from pre-existing vessels. When required, the vascular system has the potential to generate new capillary networks to maintain the proper functioning of tissues and organs. In the adult, however, angiogenesis is quite limited, occurring only in the process of wound repair and neovascularization of the endometrium during menstruation. See Merenmies, J., Parada, L.F., Henkemeyer, M., CeJJ Growth & Differenti a ti on, 8, 3-10 (1997). On the other hand, unwanted angiogenesis is the pathognomonic sign of several diseases, such as retinopathies, psoriasis, rheumatoid arthritis, macular degeneration related to age and cancer (solid tumors). Folkman, Na t ure Med., 1, 27-31 (1995). Protein kinases that are proven to be involved in the angiogenic process include three members of the tyrosine kinase growth factor receptor family: VEGF-R2 (vascular endothelial growth factor receptor 2, also known as KDR (receptor) • li1 'of the kinase insertion domain) and as FLK-1); FGF-R (fibroblast growth factor receptor); and TEK (also known as Tie-2).
VEGF-R2, which is expressed only in endothelial cells, binds to the potent angiogenic growth factor VEGF and mediates the subsequent transduction of the signal through the activation of its action as an intracellular kinase. Therefore, it is expected that direct inhibition of VEGF-R2 kinase activity will result in the reduction of angiogenesis even in the presence of exogenous VEGF (see Strawn et al., Cancer Research, 56, 3540-3545 (1996 )), as has been observed with VEGF-R2 mutants that fail to mediate signal transduction. Millauer et al., Cancer Research, 56, 1615-1620 (1996). Moreover, VEGF-R2 seems to have no other function in the adult beyond that of mediating the angiogenic activity of VEGF. Therefore, it would be expected that a selective inhibition of the kinase activity of VEGF-R2 would cause little toxicity.
Similarly, FGF-R binds to the angiogenic growth factors aFGF and bFGF and mediates the subsequent intracellular signal transduction. Recently, it has been suggested that factors of Growth such as bFGF can play a critical role in the induction of angiogenesis in solid tumors that have reached a certain size. Yoshiji et al., Cancer Research, 57, 3924-3928 (1997). Unlike VEGF-R2, however, FGF-R is expressed in a number of different cell types throughout the body and may or may not play important roles in other normal physiological processes in the adult. However, it has been reported that systemic administration of a small molecule inhibiting the kinase activity of FGF-R blocks angiogenesis induced by bFGF in mice without apparent toxicity. Mohammad et al., EMBO Journal, 17, 5996-5904 (1998).
The TEK (also known as Tie-2) is another receptor tyrosine kinase expressed only in endothelial cells that have been shown to play a role in angiogenesis. The ligation of the angiopoietin-1 factor results in the autophosphorylation of the kinase domain of the TEK and results in a signal transduction process that seems to mediate the interaction of the endothelial cells with the supporting peri-endothelial cells, facilitating the maturation of the blood vessels recently formed. The angiopoietin-2 factor, on the other hand, seems to antagonize the action of ang? Opoyetma-1 on the TEK and interrupts the angiogenesis Maisonpierre et al., Sci ence, 277, 55-60 (1997).
As a result of the aforementioned advances, it has been proposed to treat angiogenesis by the use of compounds that inhibit the kinase activity of VEGF-R2, FGF-R and / or TEK. For example, in WIPO International Publication No. WO 97/34876 it is disclosed that certain cinnoline derivatives which are inhibitors of VEGF-R2, can be used for the treatment of pathological conditions associated with abnormal angiogenesis and / or increased vascular permeability such as cancer, diabetes, psoriasis, rheumatoid arthritis, Kaposi's sarcoma, hemangioma, acute and chronic nephropathies, atheromas, arterial restenosis, autoimmune diseases, acute inflammation and eye diseases with proliferation of retinal vessels.
In addition to the above identified protein kinases, many other protein kinases have been considered as therapeutic targets, and numerous publications disclose inhibitors of kinase activity, such as the following reviews: McMahon et al., Current Opinion in Drug Di scovery and Development, 1, 131-146 (1998); Strawn et al., Exp. Opi n. Inves t. Drugs, 7, 553-573 (1998).
There is still a need, however, for other small molecule compounds that can be synthesized and are potent inhibitors of cell proliferation, for example, inhibitors of one or more protein kinases, such as CHK1, VEGF, CDK or CDK / cyclin complexes. Since CD4 can serve as a general activator of cell division in most cells, and since the complexes of CDK4 / cyclin D and CDK2 / cyclin E dominate the early phase of the Gi stage of the cell cycle, effective inhibitors are necessary and specific for CDK4 and / or CDK2 for the treatment of one or more types of tumors.
BRIEF DESCRIPTION OF THE INVENTION An object of the invention is to provide potent anti-proliferative agents.
Accordingly, an object of the invention is to achieve compounds and drug compounds that inhibit the activity of one or more kinases, such as CDK, VEGF, and CHK-1, or cyclin complexes thereof. A further objective is to provide a method effective treatment of cancer in indicated cases, through the inhibition of kinases, such as VEGF, CHK-1, CDK4 or CDK4 / cyclin complexes type D and / or CDK2 or CDK2 / Cyclin E type complexes. Another objective is to achieve pharmaceutical compositions containing compounds effective to block the transition of cancer cells towards their proliferative phase. These and other objects and benefits of the invention, which will become apparent below in the light of the detailed description, are achieved through the use of agents that control the cell cycle of the invention which will be described below.
In accordance with these objectives, Formula I represents a compound provided in accordance with the present invention.
I) where R1 is a substituted or unsubstituted alkyl, aryl, heteroaryl, carbocycle or heterocycle, or where R 4 is H or a lower alkyl, and X is an alkyl, aryl, heteroaryl, carbocycle, a substituted or unsubstituted heterocycle; and R2 is an unsubstituted or substituted alkyl, aryl, heteroapl, carbocycle, or heterocycle group, or where R 4 is H or a lower alkyl, and X is an aryl, heteroaryl, carbocycle, or substituted or unsubstituted heterocycle; or a pharmaceutically acceptable salt of a compound of Formula I; or a pharmaceutically active prodrug or metabolite of a compound of Formula I, or a pharmaceutically acceptable salt of the prodrug or metabolite.
In accordance with these objects, a compound represented in Formula II is also provided: where R '^ is an alkyl, apl, heteroa il, carbocycle, or substituted or unsubstituted heterocycle group, wherein each R4 is individually H or a lower alkyl, and X is a substituted or unsubstituted alkyl, apl, heteroapl, carbocycle, or heterocycle group; and R 'is an amino, nitro, alkenyl, alkyl, aryl, heteroaryl, carbocycle, or substituted or unsubstituted heterocycle group, where R 4 is independently H or a lower alkyl, and X is a substituted or unsubstituted apl, heteroapl, carbocycle or heterocycle group; or a pharmaceutically acceptable salt of a compound of Formula II; or a pharmaceutically active prodrug or metabolite of a compound of Formula II, or a pharmaceutically acceptable salt of the prodrug or metabolite.
Also provided in accordance with the invention is a pharmaceutical composition which includes: (a) a cell cycle control agent selected from: (i) a compound of Formula I or II, (11) a pharmaceutically acceptable salt of a compound of Formula I or II; or (m) a pharmaceutically active prodrug or metabolite of a compound of Formula I or II, or a pharmaceutically acceptable salt of the prodrug or metabolite; Y (b) a pharmaceutically acceptable carrier.
The invention also provides methods for obtaining the compounds of Formula I and II.
In addition, according to the invention, the method of using a compound as a cell cycle control agent for the treatment of diseases or disorders mediated by the inhibition of quasis, by administration to the patient who requires it, is provided composed of Formula I or II, or a salt of a pharmaceutically acceptable compound of Formula I or II; or a pharmaceutically active prodrug or metabolite of a compound of Formula I t or II, or a pharmaceutically acceptable salt of the prodrug or metabolite.
Additionally, the invention provides a method for treating fungal infections, neoplasia, or cancer, as well as other pathological conditions associated with unwanted angiogenesis and / or cell proliferation, including the administration, to a patient in need of such treatment, of effective amounts of a compound of Formula I or II or a pharmaceutically acceptable salt of a compound of Formula I or II; or a pharmaceutically active prodrug or metabolite of a compound of Formula I or II, or a pharmaceutically acceptable salt of the prodrug or metabolite.
The invention also provides a method for modulating and / or inhibiting the activity of kinases by means of administration to the patient that requires it, of a compound of Formula I or II or a salt of a compound of Formula I or II pharmaceutically acceptable; or a pharmaceutically active prodrug or metabolite of a compound of Formula I or II, or a pharmaceutically acceptable salt of the prodrug or metabolite. 25 Also provided according to the invention is a pharmaceutical composition containing a compound of Formula I or II or a pharmaceutically acceptable salt of a compound of Formula I or II; or a pharmaceutically active prodrug or metabolite of a compound of Formula I or II, or a pharmaceutically acceptable salt of the prodrug or metabolite, and the therapeutic use of the composition in the treatment of diseases mediated by the activity of kinases, such as cancer , as well as other pathological conditions associated with undesired angiogenesis and / or cell proliferation, such as diabetic retinopathy, neovascular glaucoma, rheumatoid arthritis and psoriasis.
For the pharmaceutical composition and methodological aspects of the invention, Ri may also be, hydrogen, in Formula I and II.
The agents of the invention and the compositions containing said agents may be useful in the treatment of various disorders or pathological conditions associated with uncontrolled or undesired cell proliferation, such as cancer, autoimmune disorders, viral diseases, fungal diseases, neurodegenerative disorders, and diseases. cardiovascular Therefore, the invention is also oriented to methods for the treatment of such diseases by administration of an effective amount of the agent of the invention.
Other aspects, benefits, and characteristics of the invention will become apparent from the detailed description presented below.
DETAILED DESCRIPTION AND PREFERRED EMBODIMENTS OF THE INVENTION The compounds and compositions of the present invention are useful as antiproliferative agents and as inhibitors of kinase complexes of mammals, insects and fungi. For example, the VEGF, CHK-1 and / or CDK complexes can be inhibited. Such compounds and compositions are also useful for controlling proliferation, differentiation and / or apoptosis.
Preferred Ri, R2, R'i and R'2 are exemplified below in the compounds of the groups of Formula I or 11: Preferably, Ri and R'i are Li ?? ¿IÉ¿ ^ where Y is CH or N or CR3, X is as defined above and R3 is H, or one or more substitutes located on the ring, such as an alkyl, alkenyl, aryl, heteroaryl, carbocycle, heterocycle, hydroxy, halogen, alkoxy group , aryloxy, heteroaryloxy, thioalkyl, thioaryl, thioacyl, thioheteroap or substituted or unsubstituted amino; or where the two Y can be the same or different. In these implementations, in which Ri or R 'i is there are one or more substitutes of R3 in the phenyl ring Preferably, Ri and R'i are substituted or unsubstituted where the R3 groups are as described above. In addition, two R3 together with an adjacent nitrogen can form a heteroaryl ring or heterocycle.
Preferably, R2 are substituted or unsubstituted phenyls or where R is H or a lower alkyl, and X is a substituted or unsubstituted alkyl, aryl, heteroaryl, carbocycle or heterocycle selected group.
Other preferred R2 and R 'groups are substituted or unsubstituted heteroaryls such as Other preferred groups R2 and R'2 are where R3 is as defined above Preferred substituent groups for the phenyl of R 2 include fluorine, chlorine, hydroxyl, or an alkoxy group, such as methoxy. Examples of the preferred R, X, and Y groups are found in the following exemplified compounds.
And it is preferably nitrogen.
X is preferably aryl, heteroaryl * carbocycle, or heterocycle, more preferably phenyl.
R2 and R'2 can also be an amino (-NR'R ") wherein R 'and R" are independently defined as R3 above, and together with an adjacent nitrogen can form a ring.
R 4 is preferably a hydrogen, or it may be a lower alkyl containing 1 to 6 carbon atoms, which may be substituted or unsubstituted. The two R4 can be the same or different.
Other preferred Ri, R2, R'i and R'2 groups are found in the following exemplified compounds.
Any desired alkyl group can be used, for example, as Rx or R2 or R 'i or R' 2 or R3 or X. The alkyl group can be a straight or branched chain alkyl group having one to twelve carbon atoms. Exemplified alkyl groups include methyl, ethyl, n-propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, tert-pentyl, hexyl, isohexyl, and the like. The alkyl can be substituted or unsubstituted. Preferred substituted alkyls include fluoromethyl, difluoromethyl, trifluoromethyl, 2-fluoroethyl, 3-fluoropropyl, hydroxymethyl, 2-hydroxyethyl, 3-h? droxipropil, and the like.
Any desired aryl, heteroaryl, carbocycle, or heterocycle group can be used as, for example, R% R; or R 'R-. X, Groups can be fused or non-fused, monocyclic or polycyclic.
Preferred aryl and heteroaryl groups include unsaturated or aromatic ring structures, * «*" 32 monocyclic and polycyclic, referring as "aril" to those who are carbocycles and as "heteroaryl" to those who are heterocycles. Examples of ring structures include phenyl, naphthyl, 1,2,3-tetrahydronaphthyl, furyl, thienyl, pyrrolyl, pyridyl, pyridinyl, pyrazolyl, imidazolyl, pyrazinyl, pyridazinyl, 1,2,3-triazinyl, 1, 2, 4-Oxadiazolyl, 1,3,4-oxadiazolyl, lH-tetrazol-5-yl, indolyl, quinolinyl, benzothiophenyl (tylphthanyl), furanyl, thiophenyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, triazolyl, tetrazolyl, isoquinolinyl, acridinyl, pyrimidinyl , benzimidazolyl, benzofuranyl, and the like.
Preferred carbocyclic groups include those having from three to twelve carbon atoms, including bicyclic and tricyclic cycloalkyl structures. Preferred carbocyclic groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and the like.
Preferred heterocyclic groups include saturated rings containing carbon atoms, for example rings of 4 or 5 carbon atoms and at least one heteroatom selected from nitrogen, oxygen and sulfur, and without unsaturation. Heterocyclic groups include pyrrolidinyl, piperidinyl, thiazinyl, and morpholinyl.
Ri / R2, R3 / X and other R groups can be unsubstituted or substituted with any desired substituents or substituents that do not adversely affect the desired activity of the compound. Examples of preferred substituents are those found in the following exemplified compounds, as well as halogens (chlorine, iodine, bromine, or fluorine); C? -6-alkyl; Ci-e-alkenyl; Ci- '6-alkinil; hydroxyl; C? -6-alkoxyl; Not me; nitro; thiol; thioether; imina; cyano; amido; phosphonate; phosphine; carboxyl; thiocarbonyl; sulfonyl; sulfonamide; ketone; aldehyde; ester; oxygen (= 0); haloalkyl (for example trifluoro et al); carbocyclic cycloalkyl, which may be monocyclic or polycyclic non-fused or fused (eg, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl) or a heterocycloalkyl, which may be monocyclic or fused or non-fused polycyclic (eg, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, or thiaziml); carbocyclic or heterocyclic, monocyclic or polycyclic aryl not fused or fused (for example, phenyl, naphthyl, pyrrolyl, indolyl, furanyl, thiophenyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, triazolyl, tetrazolyl, pyrazolyl, pyridinyl, quinolinyl, isoquinolinyl, acridinil, pyrazinyl, pyridazmyl, pyrimidinyl, benzimidazolyl, benzothiophenyl, or benzofuranyl); ammo (primary, secondary or tertiary); nitro; thiol; thioether; O-lower alkyl, Q- apl, aril; aryl-lower alkyl; C02CH3, CONH2; OCH2CONH :; NH2; S02NH2; OCHF2; CF3; 0CF3; and similar. Such molecules can also be optimally substituted by a fused or bridged ring structure, for example OCH2-0. These substituents can in turn be optimally substituted with a substituent selected from such groups. Preferred compounds are shown in the following examples, and also: The present invention also relates to intermediates useful in the preparation of compounds of Formula I or II. A particularly preferred intermediary has the structure Another preferred intermediary has the structure Another preferred intermediary has the structure X = halogen, N02 [X = Halogen, N02J In place of SEM, in the above-mentioned three intermediates, other known protecting groups can be used, such as benzyloxycarbonyl (CBZ), tertiary-bicarbomethyl (BOC), tetrahydropyranyl (THP), and fluoro-9-methyloxycarbonyl (FMOC) . I b Other preferred intermediaries include twenty 5 19C The abbreviations "SEM" and "PMB" refer to (trimethylsilyl) ethoxymethyl and p-methoxybenzyl, respectively. A preferred intermediary has the structure where PG is a protecting group, T is a reactive group such as boron, halogen, N02, or unsubstituted or substituted NH2, and T 'is a reactive group such as CHO, C02H, CO2R3 * CONR3R3, where the R3 groups are as define above. The pharmaceutical compositions according to the invention can comprise, as an active component, alternatively or additionally to a compound of formula I or II, a pharmaceutically acceptable salt of a compound of formula I or II, or a pharmaceutically active prodrug or metabolite of such compound or salt or a salt of the prodrug or metabolite. Such compounds, salts, prodrugs and metabolites are sometimes referred to herein together as "cell cycle control agents". The term "prodrug" refers to the metabolic precursor of a compound of Formula I or II (or a salt thereof) that is pharmaceutically acceptable. A prodrug can be inactive when it is administered to a subject, but it has not been converted into an active compound of Formula I or II. The term "active metabolite" refers to a metabolic product of a compound of Formula I or II that is pharmaceutically acceptable and effective. The prodrugs and active metabolites of compounds of Formula I and II can be determined using known techniques. The active metabolites and prodrugs of a compound can be identified using routine techniques known. See, for example, Bertolini et al., J. Med. Chem. , 40, 2011-2016 (1997); Shan, et al., J. Pharm. Sci. , 86 (7), 765-767; Bagshawe, Drug Dev. Res. , 34, 220-230 (1995); Bodor, Advances m Drug Res. , 13, 224-331 (1984); Bundgaard, Design of Prodrugs (Elsevier Press 1985); and Larsen, Design and Application of Prodrugs, Drug Design and Development (Krogsgaard-Larsen et al., eds., Har ood Academic Publishers, 1991). It is understood that within the invention a compound of Formula I or II can exhibit the phenomenon of tautomerism and that the figures of the formula with their corresponding specifications represent only one of the possible tautomeric forms. It is to be understood that the invention encompasses any tautomeric form that modulates and / or inhibits the activity of quasars and should not be limited merely to a single tautomeric form used within the figures of the formula. Some of the compounds of the invention may exist as simple stereoisomers (e.g., essentially free of other stereoisomers), racemates, and / or mixtures of enantiomers and / or diastereomers. It is intended that all of these simple stereoisomers, racemates and mixtures thereof be encompassed in the present invention. Preferably, the optically active compounds of the invention are used in an optically pure form.
As generally understood by those skilled in the art, an optically pure compound containing a chiral center (e.g., an asymmetric carbon atom), is one that consists essentially of one of the two possible enantiomers (ie, is enantiomerically pure). ), and an optically pure compound containing more than one chiral center is one that is both enantiomerically and diasterically pure. Preferably, the compounds of the present invention are used in a form that is at least 90% optically pure, ie, a form that contains at least 90% of a simple isomer (80% enantiomeric excess ("ee") or excess diastereomer ("de")), more preferably at least 95% (90% ee or de), better yet at, minus 97.5% (95% ee or de), and most preferred is at least 99% (98% ee or of). Additionally, Formulas I and II are intended to cover both the solvated and unsolvated forms of the identified structures. For example, Formulas I and II include compounds of the structure indicated in both their hydrated and non-hydrated forms. Other examples of solvates include the structures in combination with isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid or ethanolamine. "A pharmaceutically acceptable salt" is intended to mean a salt that retains the biological effectiveness of '** < ** »*, # *. the free acids and bases of the specific compound and which is not biologically or in any other undesirable form. A compound of the invention may contain sufficient acidic, basic functional groups, or both, and react with any of a number of organic or inorganic bases, and organic or inorganic acids, to form a pharmaceutically acceptable salt. Examples of pharmaceutically acceptable salts are those prepared by reacting the compounds of the present invention with a mineral or organic acid or an inorganic base, such as salts including sulfates, pyrosulfates, bisulfates, sulphites, bisulfites, phosphates, monohydrogen phosphates, dihydrogen phosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formats, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butin-1,4- dioates, hexin-1, 6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, ethoxybenzoates, phthalates, sulfonates, xylene sulphonates, phenylacetates, phenylpropionates, phenylbutyrates, citrates, lactates,? -hydroxybutyrates, glycolates, tartrates, methanesulfonates, propanesulfonates, naphthalene-1-sulfonates, fift naphthalene-2-sulfonates and mandelates.
If the compound of the invention is a desirable pharmaceutically acceptable salt base, it can be prepared with any suitable method available in the industry, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, acid sulfuric acid, nitric acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyrovic acid, oxalic acid, glycolic acid, acidole? salicylic acid, a pyranosidic acid, such as glucuronic or galacturonic acid, an alphahydroxide acid, such as citric or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid or cinnamic acid, a sulfonic acid, such as -toluenesulfonic or 15 ethanesulfonic acid, or the like. If the compound of the invention is an acid, the desirable pharmaceutically acceptable salt can be prepared by any suitable method, for example, treatment of the free acid with an inorganic or organic base, such as an amine (primary, tt, secondary or tertiary), an alkali metal hydroxide or metal hydroxide of alkali earth or the like. Illustrative examples of suitable salts include organic salts derived from amino acids, such as glycine and arginine, ammonium, primary, secondary and tertiary amines, and cyclic amines, such as piperidma, morpholine and piperacma, and salts • *. • ^ 42 inorganic derivatives of sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium. In the case of solid agents, those skilled in the art understand that the compounds and salts of the invention exist in different cpstallic or polymorphic forms, it being intended that all of them be within the scope of the present invention and specified formulas. The cell cycle control agents according to the l? invention, are useful as pharmaceuticals for the treatment of proliferative disorders in mammals, especially humans, characterized by undesired proliferation of endogenous tissues. The compounds of Formula I or II can be used for the treatment of individuals with 15 disorders associated with excessive cell proliferation, for example, cancers, psoriasis, immune disorders involving an undesired proliferation of leukocytes, and restenosis and other smooth muscle disorders. Moreover, said compounds can be used to prevent 20 de-differentiation of tissues and / or postmitotic cells. Diseases or disorders associated with abnormal or uncontrolled cell proliferation include, but are not limited to, the following: a variety of cancers, including, but not limited to, carcinoma, hematopoietic tumors of lymphoid strain, hematopoietic tumors of myeloid origin, tumors of mesenchymal origin, tumors of the central and peripheral nervous system and other tumors including melanomas, seminomas, Kaposi's sarcoma and the like. -pathological processes with characteristics of abnormal cell proliferation, for example, benign prostatic hyperplasia, familial adenomatosis, polyp, neurofibromatosis, arteriosclerosis, pulmonary fibrosis, arthritis, psoriasis, glomerulonephritis, restenosis after angioplasty or vascular surgery, hypertrophic scar formation, inflammatory disease intestinal, rejection of transplant, endotoxic shock, and fungal infections. - conditions associated with defective apoptosis, such as cancers (including but not limited to the types mentioned above), viral infections (including, but not limited to, herpes viruses, poxviruses, Epstein-Barr virus, Sindbís virus and adenovirus) , prevention of the development of AIDS in HIV-infected individuals, autoimmune diseases (including, but not limited to, systemic lupus erythematosus, rheumatoid arthritis, psoriasis, * autoimmune-mediated glomerulonephritis, inflammatory bowel disease and autoimmune diabetes mellitus), neurodegenerative disorders (which "include, but are not limited to, Alzheimer's disease, amyotrophic lateral sclerosis, retinitis pigmentosa, Parkinson's disease, AIDS-related dementia, spinal muscular atrophy , atrophy and cerebellar degeneration), myelodysplastic syndrome, aplastic anemia, ischemic insult associated with myocardial infarction, cerebrovascular accident and reperfusion injury, arrhythmia, arteriosclerosis, hepatic diseases induced by toxins or alcohol, hematological diseases (including but not limited to chronic anemia and aplastic anemia), degenerative diseases of the musculoskeletal system (including without being limited to them, osteroporosis and arthritis), aspirin-sensitive rhinosinusitis, cystic fibrosis, multiple sclerosis, kidney disease and pain from cancer. The active agents of the invention can also be useful in the inhibition of the development of invasive cancer, tumor angigogenesis and metastasis. Moreover, the active agents of the invention, for example, as inhibitors of CDKs, can modulate cellular RNA level and DNA synthesis and therefore it is expected to be useful in the treatment of viral infections such as HIV, human papilloma virus, herpes virus, Epstein-Barr virus, adenovirus, Sindbis virus, poxvirus and the like. The compounds and compositions of the invention inhibit the activity of kinases of, for example, the CDK / cyclin complexes, such as those active in the Go or Gi stage of the cell cycle, for example, CDK2, CDK4, and / or CDK6 complexes. The specific dose of a cell cycle control agent to be administered to obtain a therapeutic or inhibitory effect can be determined in a manner known in the art according to the particular circumstances of the case, including, for example, the specific agent to be administered, the route of administration, the condition that is being treated, and the individual or guest that is being treated. An example of a total daily dose of a cell cycle control agent, which can be administered in single or multiple doses, contains a dosage level of about 0.01 mg / kg of body weight to about 50 mg / kg of body weight . The cell cycle control agents of the invention can be administered by a variety of appropriate routes, such as oral, rectal, 46 Transdermal, Subcutaneous, Intravenous, Intramuscular or Transnasal "" The cell cycle control agents are preferably formulated into compositions appropriate for the desired routes of administration before being administered A pharmaceutical composition or preparation according to the invention includes an effective amount of a cell cycle control agent, optionally one or more other active agents, and a pharmaceutically acceptable carrier, such as a diluent or excipient for the agent, when the carrier serves as a diluent, it can be a solid, semi-solid, or liquid material acting as vehicle, excipient, or medium for the active ingredient (s) The compositions according to the invention can be made by mixing the active ingredient (s) with the carrier, or by dilution with the carrier, or by inclusion or encapsulation of the ingredient. active within the transporter, which can be fo capsule, sachet, paper container, or similar. Exemplary ingredients, in addition to one or more cell cycle control agents and any other active ingredient, include Avicel (microcpstamma cellulose), starch, lactose, calcium sulfate dihydrate, alba earth, sucrose, talc, gelatin, agar, pectin , acacia, magnesium stearate, stearic acid, peanut oil, olive oil, glyceryl ** monostearate, Tween 80 (polysorbate 80), 1, 3-butanediol, cocoa butter, beeswax, polyethylene glycol / propylene glycol, sorbitan monostearate, polysorbate 60, 2-octyldodecanol, benzyl alcohol, glycine, sorbic acid, potassium sorbate, disodium hydrogen phosphate, sodium chloride and water. The compositions can be prepared in any variety of forms suitable for the desired mode of administration. For example, the pharmaceutical compositions can be prepared in the form of tablets, tablets, powders, lozenges, sachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as solids or in liquid medium), ointments (for example, containing up to 10% of its weight in the form of a cell cycle control agent), soft gel and hard gel capsules, suppositories, sterile injectable solutions, sterile packaging powders, and the like. In similar fashion, the carrier or diluent may contain retarding or releasing materials known in the art, such as glycerol monostearate or glycerol distearate alone or with wax, ethylcellulose, hydroxypropylmethylcellulose, methyl merylate and the like. A variety of pharmaceutical forms can be used. Therefore, if a solid carrier, the preparation can be in the form of tablets, placed in hard gelatin capsules in the form of powder or in the form of pills or in the form of a troche or lozenge. The amount of solid carrier may vary, but will generally be from about 25 mg to about 1 g. If a liquid carrier is used, the preparation can be in the form of syrup, emulsion, soft gelatin capsule, sterile injectable solution or suspension in a vial or vial or non-aqueous liquid suspension. To obtain a stable water-soluble dosage form, a pharmaceutically stable salt of an agent of the invention is dissolved in an aqueous solution of an organic or inorganic acid such as the 0.3M solution of succinic acid or citric acid. If a soluble salt is not available, the agent can be dissolved in a suitable solvent or combinations of cosolvents. Examples of suitable co-solvents include, but are not limited to, alcohol, propylene glycol, polyethylene glycol 300, polysorbate 80, glycerin and the like in concentrations ranging from 0-60% of the total volume. A compound of Formula I or II can be dissolved in DMSO and diluted with water. The composition can also be in the form of a solution of a salt of the active ingredient in a suitable aqueous vehicle such as water or isotonic solutions of saline or dextrose. The compositions of the invention can be manufactured in commonly known forms for the preparation of pharmaceutical compositions, for example, using conventional techniques such as mixing, dissolving, granulating, drageeing, polishing, emulsifying, encapsulating, compressing or lyophilizing. The pharmaceutical compositions can be formulated in a conventional manner using one or more physiologically acceptable carriers, which can be selected from excipients and auxiliaries that facilitate the processing of the active compounds into preparations that can be used pharmaceutically. The proper formulation depends on the chosen route of administration. To be injected, the agents of the invention can be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer. For transmucosal administration, suitable penetrants are used in the formulation for the barrier to be penetrated. Such penetrants are generally known in the art. For oral administration, the compounds can be formulated simply by the combination of active compounds with pharmaceutically acceptable carriers known in the art. Such transporters allow the compounds of the invention to be formulated as tablets, tablets, dragees, capsules, liquids, gels, syrups, watered pastes, suspensions and the like, for oral ingestion by the patient to be treated. Pharmaceutical preparations for oral use can be obtained by using a solid excipient in admixture with * the active ingredient (agent), optionally grinding the resulting mixture, and processing the mixture of granules after the addition of suitable auxiliaries, if desired, to obtain tablets or dragees Suitable excipients include: fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol and cellulose preparations, eg, corn starch, wheat starch, rice starch, potato starch, gelatin, gum, hydroxypropyl ethyl -cellulose, sodium carboxymethylcellulose, methyl cellulose, or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as, linked polyvinyl pyrrolidone, agar, or alginic acid or salts thereof such as sodium alginate. Dragee centers are provided with * appropriate wraps. For this purpose, they can be use sugar solutions, which can optionally ^ >; contain gum arabic, polyvinyl pyrrolidone, g l? * Carbopol, polyethylene glycol, and / or titanium dioxide, •. 'Lacquer and organic solvents or mixture of suitable solvents. *,' * Can you add tinctures or pigments to tablets or dragee wraps for the identification or characterization of the different combinations of active agent J. The pharmaceutical preparations that can be used orally include gelatin "push-fit" capsules, as well as sealed soft capsules, made of gelatin and a plasticizer, such as glycerol or sorbitol.The "push-fit" capsules can contain the active ingredients attached by mixing to fillings such as lactose, binders such as starches, and / or lubricants such as talc or magnesium stearate, and, optionally, stabilizers In soft capsules, the active agents can be dissolved or suspended in suitable liquids, such as fatty oils , liquid paraffin, or liquid polyethylene glycol Additionally, stabilizers can be added All formulations for oral administration they should be in appropriate doses for such administration. For oral administration, the compositions can take the form of tablets or lozenges formulated in conventional manner.
For nasal or inhalation administration, the compounds for use according to the present invention are conveniently administered in an aerosol presentation form presented in pressurized or nebulized containers, by the use of a suitable propellant, for example, dichlorodifluoromethane, trichlorofluororaethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosing unit can be determined by the provision of a valve that manages an established amount. Capsules and gelatin cartridges for use in an inhaler or insufflator or the like can be formulated containing a powder mixture of the compound and a suitable powder base such as lactose or starch. The compounds can be formulated for injectable parenteral administration, for example, by bolus injection or continuous infusion. The injectable formulations can be presented in the form of metered units, for example in ampoules or multi-dose containers, with the addition of a preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulated agents such as stabilizing and / or dispersing suspensors.
Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in a water-soluble form. Additionally, suspensions of the active agents can be suitably prepared in the form of oily suspension injections. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic esters of fatty acids, such as ethyl oleate or triglycerides or liposomes. Aqueous injectable suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol or dextran. Optionally, the suspension may also contain appropriate stabilizers or agents that increase the solubility of the compounds to allow the preparation of highly concentrated solutions. For ocular administration, the active agent is presented in an acceptable pharmaceutically acceptable ophthalmic vehicle, so that the compound is kept in contact with the ocular surface for a sufficient period of time to allow the compound to penetrate the cornea and inner regions of the eye. , including / for example, the anterior chamber, the posterior chamber, the vitreous body, the aqueous humor, the vitreous humor, the cornea, the iris / ciliary body, the lens, the choroid / retina and the sclera.
The pharmaceutically suitable ophthalmic vehicle may be an ointment, vegetable oil or encapsulating material. The compound of the invention can also be injected directly into the vitreous and aqueous humor. Alternatively, the active ingredient may be in powder form to be reconstituted prior to use with an appropriate vehicle, eg, sterile, pyrogen-free water. The compounds can also be administered rectally in compositions such as suppositories or retention enemas, for example, containing conventional suppository bases such as cocoa oil or other glycerides. The compounds can also be formulated in depot preparations. Such long-acting formulations can be administered by implantation (e.g., subcutaneous or intramuscular) or by intramuscular injection. For this reason, the compounds can be formulated, for example, with suitable polymeric or hydrophobic materials (as an emulsion in an acceptable oil) or with ion exchange resins, or as sparingly soluble derivatives, for example, a sparingly soluble salt. A pharmaceutical carrier for hydrophobic compounds is a solvent system containing benzyl alcohol, a non-polar surfactant, an organic polymer miscible with water and an aqueous phase. The soldering system can be a VPD co-solvent system. VPD is a solution of 3% w / v of benzyl alcohol, 8% w / v of non polar polyester surfactant 80 and 65% w / v of polyethylene glycol 300, brought to volume in absolute ethanol. The VPD co-solvent system (VPD: 5W) contains VPD diluted 1: 1 with a dextrose solution in 5% water. This cosolvent system dissolves hydrophobic compounds well, and by itself produces little toxicity when administered systemically. Naturally, the proportions of a system The dissolvent can be varied considerably without destroying its solubility and toxicity characteristics. Moreover, the identity of the components of the cosolvent can be varied. For example, other non-polar low-toxicity surfactants may be used instead of polysorbate 80; the fractional size of polyethylene glycol can be varied; polyethylene glycol can be replaced by other polymers • biocompatible, for example, dextrose can be replaced by polyvinylpyrrolidone and other sugars or polysaccharides. Alternatively, other hydrophobic pharmaceutical compound delivery systems may be used. Liposomes and emulsions are examples, known from release vehicles or transporters for hydrophobic drugs. Certain organic solvents such as dimethylsulfoxide can also be used, although usually with the i. '* * and * X - 56 cost of greater toxicity. Additionally, the compounds can be administered using a sustained release system, in the form of semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. 5 Various sustained release materials have been established and are known to those skilled in the art. Sustained-release capsules can, depending on their chemical nature, release the compounds for a few weeks and up to more than 100 days. According to the In the chemical nature and biological stability of the therapeutic reagent, additional strategies can be employed for protein stabilization. The pharmaceutical compositions may also include suitable carriers or excipients with base 15 solid or gelled. Examples of such carriers are calcium carbonate, calcium phosphate, sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycol. Some of the compounds of the invention can be provided in the form of salts with pharmaceutically compatible counter ions. The pharmaceutically compatible salts can be formed with many acids, including hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. The salts tend to be • $ & ** & ? 57 more soluble in aqueous solvents or other protonic solvents than the corresponding free base forms. A pharmaceutical composition according to the invention includes a cell cycle control agent and, optionally, one or more other active ingredients, as a known antiproliferative agent that is compatible with the cell cycle control agent and suitable for the indication to be treated. The compounds are useful as agents 10 antiangiogenic and as modulating agents and / or inhibitors of the activity of protein kinases, therefore provide treatment for cancer or other diseases associated with cell proliferation mediated by protein kinases. Therapeutically effective amounts of the agents of the invention can be used to treat diseases mediated by the modulation or regulation of protein kinases. An "effective amount" means the amount of an agent that, when administered to a mammal 20, which requires such treatment, is sufficient to effect the treatment of a disease mediated by the activity of one or more kinases. Thus, for example, a therapeutically effective amount of a compound of Formula I or II, salt, active metabolite or derivative prodrug is a 25 enough to modulate, regulate, or inhibit the activity of one or more kinases in such a way that a pathological condition that is mediated by that activity s, * • * reduces or is alleviated. i "Treat" means at least the mitigation of 5, a pathological condition in a mammal, such as a human, that is affected at least in part, by the activity of one or more qumases, and includes: preventing the occurrence of the pathological condition in a mammal, particularly when the mammal is predisposed to have the pathological condition but has not yet been diagnosed; modulate and / or inhibit the pathological condition; and / or alleviate the pathological condition. The agents of the invention can be prepared using the reaction routes and synthetic schemes described below, employing the techniques available in the art and using materials that are readily available. The general schemes exemplified 1 to 6, shown below, can be used to make the compounds of the invention.
Track 1 X = CI I C D E F [Pd catalyst = Pd Catalyst] The halogenated intermediate A can be obtained by standard 5-amino nomdazole diathozation and treatment of the resulting diazonium salt with an appropriate halide salt, such as CuCl or Kl. Subsequent halogenation to obtain 3-halomdazole B is achieved by treatment with a suitable base such as sodium or potassium mdroxide and an elemental halogen such as iodide. The intermediate B is protected using any number of suitable protecting groups and is treated with an alkyl or aryl boronic acid (preferably stoichiometric) or ester and a suitable Pd catalyst, for example, Pd (PPh3), to affect the selective reaction in the C-3 position. A subsequent reaction with a second alkyl or aryl boronic acid or ester and a suitable Pd catalyst makes it possible to obtain the desired intermediate substance 3, 5-d? Sust? E?, Which is then deprotected to obtain the final compound F.
The conditions of deprotection are consistent with Q * -specific protective group used, for example-acid conditions for the removal of a protective group, ** THP. R; and R2 are as defined above and can be R ',, and R'2. Variation of track 1 [Pd catalyst = Pd Catalyst] The alternative synthetic variation to lane 1 presented above requires the treatment of intermediate C, where X is Cl with alkyl dithm species, such as hexamethyl dithm, and an appropriate Pd catalyst, to obtain the intermediate substance G. The reaction of intermediate G with an alkyl or aryl halide and an appropriate Pd catalyst provides the desired intermediate D which may continue to be worked up as described above.
Via 2 *.
[Pd catalyst = Catalyst, Pd Diammoethane = Diaminoethane] Alternatively, as shown in lane 2 above, a 5-n? Tromdazole can be halogenated as described above for intermediate A, to obtain a nitro compound H, by treatment with a b ^ se suitable as sodium hydroxide or potassium hydroxide and an elemental halogen as the iodide to obtain an intermediate substance I after the standard protection cbn a suitable protective group. With the treatment of intermediate I with alkyl dithm species, such as ditin hexamethyl and a suitable Pd catalyst, intermediate substance J can be obtained. A further reaction of the nitro compound J with an alkyl or aryl boronic acid or ester and a suitable Pd catalyst makes it possible to obtain the 3-substituted-K mdazole. The amine is obtained by reducing K with a suitable reducing agent, such as hydrogen with palladium catalyst or SnCl2. The diazotization of the resulting 5-amino indazole and the treatment of the resulting diazonium salt with a salt of Suitable halide, such as CuCl or Kl, produces the intermediate substance with halo L. The reaction of L with an alkyl or aryl boronic acid or ester and a suitable Pd catalyst produces the intermediate M which is deprotected as before to give the final compound F. Rx, and R are those defined above $ and can be R 'i and R'2. ID " 5 : > ^, - 63"'PA [Acid activation« = Acid activation / Protécti n - r * Protection, Reduction = Reduction] In lane 3 shown above, a B - carboxy - indazole is activated to provide an active acyltin species, such as carbonyl di-imidazole, which is treated with a suitable alkoxy alkyl amine, such as N, N-dimethylhydroxylamine, resulting in amide A 'The selective halogenation of intermediate A' j with an elemental halogen such as bromine or iodine, and preferably with a catalyst such as bis (trifluoroacetoxy) yodosobenzene or bis (acetoxy) iodosobenzene produces indazole B ', of 5 halos Protection of intermediate B' under standard conditions with an appropriate protecting group as PMB or THP provides a protected C-amide.The reduction of C with an appropriate reductant such as lithium aluminum hydride or an equivalent hydride reducing agent provides the key intermediate aldehyde D '. The R3 is corao defined above, pref. preferably an alkyl, substituted or unsubstituted, preferably a lower alkyl.
Track 4 ; Deprotect? On = Deprotection] In path 4 shown above, intermediate D 'reacts with a substituted "B" diamine and a suitable oxidizing agent such as sulfur to provide benzimidazole C ". The conversion of compound C "to the corresponding borated ester D" is achieved by reacting with a suitable diboro species, such as dipychocholate or other boron electrophilic source, with a suitable palladium catalyst. Intermediate D "then reacts, under a palladium catalyst, with a halogenated alkyl or aryl halide to give substituted 5-indazole, intermediate E", which after an appropriate deprotection provides the final compound H. "Alternatively, the initial compound D 'is reacted with a suitable diboro species, such as the bis (pinacolato) diboro or another suitable electrophilic boron source and a palladium catalyst suitable for giving the boron ester F ". The preparation of compound F "to intermediate D" is achieved as described above for intermediate D '. Another alternative conversion can be achieved by the reaction of the intermediate aldehyde F "with an alkyl or substituted aryl halide to provide R2 with a palladium catalyst for provide G "which then further reacts with a substituted" B "diamine and a suitable oxidizing agent, such as sulfur, to provide the benzimidazole E." As before, deprotection allows to obtain the final compound H ". R2 is as defined above and can be R'2. R3 is as defined above. A further preparation of the intermediate E "can be achieved by acid or alkyl borinic ester in a suitable palladium catalysis." Other electrophilic boron species that can be used in the structure: R3 R3 B-B where R3 is as defined above and R3 groups can form a ring R3? ' OR3 [Where R3 as defined above and two R3 groups can form a ring. = Where R3 is as defined above and two R3 groups can form a ring.] Specific examples include: Track 5 [Deprotection = Deprotection] In lane 5 above, the intermediate alcohol X: can be activated, for example, by reaction with a sulfonyl halide, such as methanesulfonyl chloride and a suitable base such as triethylamine, and this electrophilic species then it reacts with a nucleophile, such as a substituted amine, to provide the intermediate substance X2 which is then deprotected under the appropriate conditions. R2 is as defined above, and may be R'2. R3 is as defined above. \ ' '4 69 final products. 'Ri and R2 are as defined above and can be R'i and R'2. The preparation of the specific preferred compounds of the invention is described in detail in the following examples. The artisan will recognize that the described chemical reactions can readily be adapted to prepare a number of other inhibitors of kinases of the invention. For example, the synthesis of compounds not exemplified according to the invention can be carried out I0 .1t- successfully through modifications known to those who are experts in the art, for example, by means of the appropriate protection of interfering groups, the exchange for other suitable reactants known in the art or the modification in the routine conditions of the reaction. As an alternative, other reactions disclosed herein or known in the art will be recognized that are applicable for the preparation of other compounds of the invention. In the examples described below, except where indicated, all temperatures are presented at 0 degrees Celsius and all parts and percentages are by weight. The reactants were purchased from commercial suppliers such as Aldrich Chemical Company or Lancaster Synthesis Ltd. and were used without further purification unless otherwise indicated. Tetrahydrofuran (THF) 5 distilled from calcium hydride and N, N-dimethylformamide (DMF) were purchased from Aldrich in sealed "Sure seal" bottles and used in the way they were received. All solvents were purified using standard methods known to those skilled in the art, unless otherwise indicated. The reactions indicated below were generally made under a positive argon pressure or with a drying tube, at room temperature (unless otherwise clarified), in anhydrous solvents, and the reaction vessels were provided with rubber holes for the introduction of substrates and reagents with a syringe. The glass elements were dried in the oven and / or by heat. Analytical thin-layer chromatography (TLC) was performed on Analtech 60 F 254 glass-silica gel plates (0.25 mm), extracted with solvents in appropriate proportions (v / v) and denoted where appropriate. The reactions were tested by TLC and finished judging by the consumption of the initial material. The visualization of the TLC plates was carried out with a p-anisaldehyde reagent spray or a phosphomolybdic acid reagent (Aldrích Chemical 20% by weight in ethanol) and activated with heat. The procedures were typically done by doubling the reaction volume with the reaction or extraction solvent and then washing with the indicated aqueous solutions using 25% by volume of extraction volume unless otherwise indicated. The product solutions were dried over Na2SO4 or anhydrous MgSO4 prior to filtration and evaporation of the solvents under reduced pressure in a * x Rotary evaporator and annotated as solvents removed in vacuo. 3te performed flash column chromatography (Still et al., J. Org. Chem., 43, 2923 (1978)) using flash silica gel with Baker graduation (47-61 μm) and a silica gel: material ratio crude from approximately 20: 1 to 0 50: 1, unless otherwise indicated. The hydrogenation was carried out at the pressure indicated in the examples or at ambient pressure. The 1 H-NMR spectra were recorded on a Bruker instrument operating at 300 or 500 MHz and the 5 13 C-NMR spectra were recorded operating at 75 MHz. The NMR spectra were obtained as CDC 13 solutions (reported in ppm), with chloroform as reference standard (7.25 ppm and 77.00 ppm), CD3OD (3.4 ppm and 4.8 ppm and 49.3 ppm) or internal tetramethylsilane (0.00 ppm) when appropriate. Other NMR solvents were used as needed. When the maximum multiplicities are reported, the following abbreviations are used: s (smgleta), d (doublet), t (triplet), m (multiplet), br (extended), dd (doublet of doublets), dt (doublet of triplets ). When they occur coupling constants, they are expressed as hertz (Hz). The infrared (IR) spectra were recorded 'in. . * pg ..; í an Perkin-Elmer FT IR spectrometer as clean oils or KBr pellets, and are reported in wave numbers (c "1) .. Mass spectra were obtained using LSIMS or electrospray. All melting points (mp) are uncorrected The starting materials used in the examples are commercially available and / or can be prepared with techniques known in the art.
Example 1: 5-Phenyl-3-Est r? L-lfí-Indazol [dioxane = dioxane, Toluene = Toluene, reflux = reflux] (a) Intermediate substance la-5-Chloro-3-yada-lH-mdazole: 5-Am? no-lH-mdazole (15.41 g, 116 mmol) 4 se suspended in a mixture of water (250 ml), ice (250 ml), and concentrated HCl (100 ml). The mixture was cooled in an ice bath and salt to an internal temperature of -5 ° C. S. added to this mixture a solution of sodium nitrite (8.78 g, 127 mmol) in water (75 ml), which had been cooled to 0 ° C. The resulting diazomo solution was stirred for 15 minutes at -5 ° C. copper chloride solution (I) { 14 .9 "'vtti g, 151 mmol) in concentrated HCl (150 ml) was cooled to 0 ° C and then added dropwise to the diazonium solution," - causing the formation of an orange precipitate.The cold bath was then removed To allow the reaction to warm to room temperature, the evolution of the gas started at an internal temperature of 10 ° C. After stirring at room temperature for 1.5 hours, the evolution of the gas gave way, then the flask was heated to 60 ° C for 30 ,. \ 6 minutes and then cooled to ~ 15 ° C. A brown precipitate formed. The precipitate was collected by suction filtration and dried in a tumble dryer over NaOH for 16 hours to give crude 5-chloro-1-indazole (25.6 g) as a tan powder. 15 This crude intermediate was dissolved in 1,4-dioxane (400 ml). Aqueous 3 M NaOH (400 mL) and iodide flakes (35.3 g, 139 mmol) were added to the solution. After stirring at room temperature for 2 hours, the reaction mixture was neutralized to pH = 6 with 20% aqueous citric acid, causing the change from dark to light green * Saturated aqueous sodium thiosulfate was added (~ 400 ml) to the solution, causing the color change from green to yellow, and the solution was extracted with ethyl acetate (3 x 1000 ml). The combined organic extracts were dried over sodium sulfate, filtered by suction through coarse frit. ** t! i? £: *? .. "ß * i: ffi? £ i! üíJ2ia and concentrated to a green slurry which was then dissolved in ethyl acetate (500 ml), filtered through a pad of Celite and concentrated to a green solid. Purification by chromatography on silica gel (25% ethyl acetate in hexanes) provided 5-chloro-3-iodo-lH-mdazole (14.18 g, 44% of 5-amino-1H-mdazole) in the form of a whitish solid: mp * 198-199 ° C; Rf = 0.53 (50% ethyl acetate / hexanes); XH NMR- (DMSO-de) 6 7.44 (m, 2H), 7.60 (d, ÍH, J = 8.7 Hz), 13.68 (á, ÍH). Analysis (C7H4C1IN2) C, H, N. (b) Intermediate Ib-5-Chloro-3-? Odo-l- (2- (trimethylsilyl) -ethoxymethyl] -lH-indazole: 5-Chloro-3- was dissolved iodide-liya-indazole (8.86 g, 31.8 mmol) in THF (100 ml) and cooled in a water bath and salt at 0 ° C. Solid sodium t-butoxide (3.67 g, 38.2 mmol) was added and The mixture was stirred at 0 ° C for 1 hour, then 2- (trimethylsilyl) ethoxymethyl chloride (7.96 g, 38.2 mmol) was added, and stirring was continued at 0 ° C for an additional 1 hour. ethyl acetate (200 ml) and washed with water (100 ml) and brine (100 ml) The organic layer was dried over magnesium sulfate, filtered and concentrated chromatography on silica gel (5 to 20% acetate ethyl acetate in hexanes) gave Ib (9.75 g, 75%) as a yellow oil: Rf = 0.39 (5% ethyl acetate / hexanes); XH NMR (CDC13) d -0.06 (s, 9H ), 0.87 (t, f 1 »* * '. 76 2H, J = 8.1 Hz), 3 # 55 (t, 2H, J = 8.1 Hz), s5.70 (s, 2H), 7.43 (dd, ÍH, J = 8.9, 1.7 Hz), 7.49 (m, 2H ). Analysis > (C? 3H? 8ClIN2OSÍ) C, H, N. (c) Intermediate le-5-Chloro-3-styryl-l- [2- (trimethylsilanyl) -ethoxymethyl-1H-indazole: 5-Chloro-3 was dissolved -Iodo-2-SEM-indazole Ib (553 mg, 1.35 mmol), styrylboronic acid (300 g, 2.03 mmol) and tetrakis (triphenylphosphino) palladium (78.2 mg, 0.068 mmol) nf toluene (10 ml) and methanol (1.4 ml) ). A saturated aqueous solution of sodium bicarbonate (1.7 ml) was added and the mixture i was heated in an oil bath at 90 ° C for 3 hours. A slight reflux was observed. After cooling to room temperature, the solution was diluted with water (15 ml) and extracted with ethyl acetate (4 x 50 ml). The combined organic extracts were dried over magnesium sulfate, filtered, and concentrated. Purification by chromatography on silica gel (toluene) provided pure lc (350.7 mg, 67%) as a yellow-colored oil: Rf = 0.20 (toluene); XH NMR (CDC13) d -0.09 (s, 9H), 0.86 (t, 2H, J = 8.1 Hz), 3.55 (t, 2H, J = 8.3 Bz), '> 5.65 (s, 2H), 7.2-7.4 (m, 7H), 7.54 (d, 2H, J = 7.6.H2), and 7.93 (d, ÍH, J = 1.6 Hz). 13C NMR (CDC13) d -1.5, 17.7, 66.5, 77.9, 111.0, 119.2, 120.3, 123.6, 126.5, 127.3, 127.4, '128.0, 128.7, 131.6, 136.9, 139.4, 142.5. Analysis (C2? H2SClN2OS? »0.02 CHC13) C, H, N, Cl. (d) Intermediate substance Id-5-Phenyl-3-styryl-l- [2- (triraethylsilanyl) -ethoxymethyl] -lfi-indazole: To a solution of 5-chloro-3-styryl-2-SEM-in? jtezbl _ lc (209.4 mg, 0.544 mmol) in dry 1,4-dioxan (0.5 ml) Be added phenylboronic acid (69.6 mg, 0.571 mmol), cesium carbonate (213 mg, 0.653 mmol), and tris (dibenzylidinacetone) dipalladium ( 10.0 mg, 0.0108 mmol). A solution of tri-tert-butyl phosphino in 1,4-dioxane (0.1 M, 0.217 ml) was added and the mixture was heated at 80 ° C for 6 hours. After cooling to room temperature, the solution was diluted with ethyl ether (20 ml), and filtered through a pad of Celite to remove the black precipitate of palladium. The filtrate was dried over magnesium sulfate, filtered, concentrated and purified by chromatography on silica gel (toluene) to give ld (77.2 mg, 33%) as a colorless oil: Rf = 0.09 (toluene); XH NMR * (CDC13) d -0.04 (s, 9H), 0.93 (t, 2H, J = 8.1 Hz), 3.62 (t, 2H, J = 8.1 Hz), 5.76 (s, 2H, J), 7.3- 7.7 (m, 14H) 8.17 (s, ÍH). Analysis (C27H3oN2OSi «0.2 H20) C, H, N. (e) Example 1 - 5-Phenyl-3-Styryl-lH-Indazole: Intermediate ld (68.1 mg, 0.16 mmol) was dissolved in absolute ethanol (2.0 ml ) and 3 M HCl (2.0 ml). The solution was heated to reflux for 20 hours, cooled to room temperature, and extracted with ethyl acetate (3 x 30 ml). The organic extracts The combined extracts were dried over magnesium sulfate, filtered, concentrated and purified by chromatography on silica gel (25 to 50% ethyl acetate in hexanes), which yielded the title compound (19.2 mg, 40%) in the form of a white solid: R: - = 0.14 (25% ethyl acetate / hexanes); XH NMR (CDC13) d 6.92 (d, 1H, J = 6.3 Hz), 7.3-7.7 (m, 13H), 8.20 (s, ÍH), 10.3 (br S, ÍH). HRMS calculated for C2: HnN2 297.1392 (MH +), found 297.1398. Analysis (C2? H? 6N2-0.7 H20) C, H, N.
Example 2: 3,5-D st r? L-lH-Indazol i. (a) Intermediate 2a-5-iodo-lfí-indaz © l: 5-Amino-lf-indazole (10.21 g, 76.7 mmol) was suspended in a mixture of water (100 ml), ice (100 ml), and Concentrated HCl (35 ml). The mixture was cooled in an ice bath and salt to an internal temperature of -5 ° C. To this mixture was added a solution of sodium nitrite (5.82 g, 84.4 mmol) in water (30 ml), which had been cooled to 0 ° C. The resulting diazonium solution was stirred for 10 minutes at -5 ° C, then a solution of potassium iodide (15.3 g, 92 mmol) in water was slowly added dropwise (50 ml). foam with the first drops of the Kl solution, and then a black, tarred gum was formed.After the addition was completed, the mixture was heated at 90 ° C for 1 hour.The tarry precipitate dissolved and a purple vapor evolved during heating, then the reaction was cooled to room temperature, causing the "formation of a fine brown precipitate." This precipitate was collected by suction filtration, and dried under vacuum to provide 5-iodoindazole 2a (14.12 g, 15%.) as a brown powder: Rf = 0.28 (50%). ethyl acetate / hexanes); XH NMR (DMSO-d6) d 7.40 (d, 1H, J = 9.0 Hz), 7.56 (dd, ÍH, J = 8.5, 1.5 Hz), 8.01 (s, ÍH) 8.16 (s , 1H), 13.23 (s, ÍH) Analysis (C7H5IN2) C, H, I, N. (b) Intermediate 2b-3,5-Diiodes-lf-indazole: Intermediate 2b was prepared by a synthetic method analogous to that of the synthesis of the intermediate substance la. The treatment of intermediate 2a with iodide and sodium hydroxide afforded 3,5-diiodo-lH-mdazole 2b (84%) in the form of a yellow solid: Rf = 0.39 (30% ethyl acetate / hexanes); XH NMR (DMSO-d6) d > < 7.41 (d, ÍH, J = 8.7 Hz), 7.66 (dd, ÍH, J = 8.7, 1.5 Hz), 7.77 (d, ÍH, J = 0.9 Hz) 13.65 (s, ÍH). (c) Intermediate 2c-3,5-Diiodo-l- [2- (trimethylsilanyl) -ethoxymethyl] -lfi-indazole: By a synthetic method similar to that of intermediate Ib, the treatment of 3,5-diiodoindazole 2b with sodium t-butoxide and 2- (trimethylsilyl) ethoxymethyl chloride gave 2c (64%) as a yellow oil: Rf = 0.53 (30% ethyl acetate / hexanes); 1H NMR (CDC13) d -0.05 (s, 9H), 0.86 (t, 2H, J = 8.1 Hz), 3.54 (t, 2H, J = 8.1 Hz), 5.69 (s, 2H), 7.34 (d, HI) , J = 8.7 Hz), 7.69 (dd, ÍH, J = 8.7, 1.5 Hz), 7.87 (d, ÍH, J = 1.5 Hz). (d) Intermediate 2d-3,5-Distryl-l- [2- (trimethylsilanyl) -ethoxymethyl] -lH-indazole: Styrylboronic acid (186 mg, 1.26 mmol) was added to the solution of 2c (210.0 mg, 0.42 mmol) and tetrakis (triphenylphosphino) palladium (48.5 mg, 0.042 mmol) in toluene , (3.5 ml) and methanol (0.5 ml). A saturated aqueous solution of sodium bicarbonate (1.05 ml) was added, and the mixture was heated in an oil bath at 90 ° C (slight reflux) for 4 hours. After cooling to room temperature, the reaction was poured into water (15 ml) and extracted with ethyl acetate (4 x 50 ml). The combined organic extracts were dried over magnesium sulfate, filtered / concentrated and purified by chromatography on silica gel (toluene) to give 2d (170.9 mg, 90%) as a yellow oil. :, Rf = 0.10 (toluene); XH NMR (CDC13) d 0.01 (s, 9H), 0.98 (t, 2H, J = 8.5 Hz), 3.67 * (t, 2H, J = 8.5 Hz), 5.73 (s, 2H), 7.17 (d, 1H, J - 16 Hz), 7.3-7.7 (m, 15H), 8.05 (s, 1H). 13C NMR ÍCDCI3) d -1.5, 17.6, 66.4, 77.7, 110.1, 119.4, 119.8, 123.3, 125.1, 126.3, 126.5, * 127.8, 128.6, 128.7, 128.9, 131.3, 137.1, 137.3, 140.6, • 143.3. Analysis (C29H32N2OSi »0. ICHCl3) C, H, N. (e) Example 2 - 3,5-Distiryl-lH-Indazole: Example 2 was prepared in a similar manner to Example 1, by treatment of 2d with 3M HCl, 3, 5-d-Stiryl-1 -indazole (33%) as a bright yellow solid: Rf = 0.11 (25% ethyl acetate / hexanes); XH NMR (CDCl 3) d 7.2-7.7 (m, 16H) 8 \ 076 (s, ÍH), 10.05 (br s, ÍH). HRMS calculated for C23H? 9N2 323.1548 (MH +), found 323.1552. Analysis (Cg3H18N2'0.5 H20) C, H, N. 4 & Example 3: 3- (lH-Benzoimidazole-2-? L) -S-Phenyl-lH-Indazole 3a 3b [Reflux = reflux, Toluene - toluene, Dioxane = dioxane] (a) Intermediate 3a-l- [2- (Trimethylsilyl) -ethoxymethyl] -1H-benzoimidazole: (See Whitten et al, J. Org. Chem. 51, 1891 (1986) incorporated herein by reference, with a similar procedure) . Solid lff-benzoimidazole (30 g, 254 mmol) was added in small portions to a suspension of sodium hydride (10.2 g of 60% dispersion in mineral oil, 254 mmol) in DMF (350 ml) at room temperature. The mixture was stirred for 3 hours, and then cooled to 0 ° C in a bath of ice. 2- (Trimethylsilyl) ethoxymethyl chloride (46.57 g, 279 mmol) was added dropwise over 10 minutes. The reaction was stirred for 16 hours, warming to room temperature when the ice was melted, then poured into water (11) and extracted with ethyl acetate. The organic layer was washed with brine, dried over sodium sulfate, filtered, concentrated and purified by chromatography on silica gel (50 to 85% ethyl acetate in hexanes) to provide 3a (56.63 g, 90%). in the form of an amber oil: .Rf = 0.40 (50% ethyl acetate / hexanes); XH NMR (CDC13) d - 0.04 (s, 9H), 0.90 (t, 2H, J = 8.1 Hz), 3.50 (t, 2H, »8.1 Hz), 5.53 (s, 2H), 7.31 (m, 2H) , 7.54 (m, ÍH), 7.81 (m, ÍH), 7.96 (s, ÍH). Analysis (C? 3H2oN2OSI • 0.5H20) C, H, N. 15 (b) Intermediate 3b-2-Iodo-l- [2- (tnmethylsilanyl) -ethoxymethyl] -lif-benzoimidazole: A solution of N-SEM -benzimidazole (intermediate 3a) (19.19 g, 77.25 mmol) in dry ethyl ether (150 ml) and cooled to -78 ° C in a dry 2ft ice bath / acetone, was added via drip via cannula to a solution of -butyl lithium (46 ml of 2.5 M in hexanes, 116 mmol) in dry ethyl ether (150 ml), also cooled to -78 ° C in a dry ice / acetone bath. The addition of the benzimidazole solution took 10 minutes. The agitation continued 15 25 more minutes, and during that time a color developed - M '., &' - "* * • 84 dark red. The resulting aryl lithium solution was * - * added via drip via cannula to a solution of iodide flakes (49 g, 193 mmol) in dry ether (500 ml), in turn "? \ Cooled to -78 ° c in a dry ice / acetone bath, then * ^ 5 to complete the aggregate (10 minutes), the cold bath was removed, and the reaction of the mixture was heated for 30 minutes to reach an internal temperature of - 10 ° C. Water (250 ml) was added and the mixture was washed with a saturated aqueous solution of sodium bisulfite (2 x 200 ml).; The organic layer was dried over sodium sulfate, filtered, concentrated and purified by chromatography on silica gel to provide 3-iodo-n-SEM-benzimidazole 3b (22.84 g, 80%) in the form of a sodium solid. yellow color: mp * 60-63 ° C; Rf = 0.70 (ethyl acetate); XH NMR (CDC13) d -0.04 tí (s, 9H), 0.92 (t, 2H, J = 8.1 Hz), 3.58 (t, 2H, J = 8.1 Hz), 5.53 (s, 2H), 7.27 (m, 2H), 7.51 (m, ÍH), 7.73 (m, 1H). HRMS calculated for C? 3H19IN2OS? Na 397.0209 (MNa +), found 397.0204. Analysis (C? 3H? 9IN2OSi) C, H, I, N. (c) Intermediate 3c-5-Chloro-l- [2-ft (t-imethylsilanyl) -ethoxymethyl] -3- (trimethylstanyl) -lH-indazolol : A mixture of intermediate Ib (6.25 g, 15.3 mmol), hexamethylditin (10.2 g, 30.5 mmol), and bis (tpfen? L phospho) palladium (II) dibromide (242 mg, 0.306 mmol) in toluene (50 mi) was heated to reflux for 30 minutes, then cooled, filtered, and concentrated. The i < * • > * 85 purification by chromatography on silica gel (5 to 50% ethyl acetate in hexanes) provided 3c (6.34 g, 93%) as a slightly yellow oil: Rf = 0.21 (5% ethyl acetate / hexanes), Rf = 0.23 (toluene); : H NMR (CDC13) d -0.06 (s, 9H), 0.56 (s with small sidebands, 9H), 0.87 • (t, 2H, J = 8.4 Hz), 3.54 (t, 2H, J = 8.4 Hz) , 5.75 (s, 2H), 7.34 (dd, 1H, J = 8.7, 1.8 Hz), 7.51 (d, 1H, J = 8.7 Hz), 7.66 (d, ÍH, J = 1.8 Hz). Analysis (C? 6H27ClN2OSiSn) C, H, Cl, N. (d) Intermediate 3d-5-Chloro-l- [2- (trimethylsilanyl) -ethoxy? Methyl] -3-. { 1- (2- (trimethylsilyl) -ethoxymethyl] -lfi-benzoimidazol-2-yl] -lfi-indazole: A mixture of 3c (4.47 g, 10.03 mmol), 3b (4.12 g, 11.03 mmol), tetrakis (triphenylphosphmo) palladium (0) (579 mg, 0.50 mmol), and copper (I) iodide (190 mg, 1.00 mmol) in THF i (100 ml) was heated to reflux for 1 hour.Additionally, a catalyst (580 mg, 0.50 g) was added. mmol) and Cul (200 mg, 1.05 mmol), and the reflux continued for 20 hours.After cooling to room temperature, the black precipitate was filtered, the filtrate was concentrated, and the residue was purified by gel chromatography. silica (toluene) to give pure 3d (2.29 g, 43%) as a colorless oil that crystallizes upon settling: mp = 80-82 ° C; Rf = 0.12 (10% ethyl acetate / hexanes), Rf = 0.13 ( toluene);? H NMR (CDC13) d -0.15 (s, 9H), "-0.06 (s, 9H), 0.85 (t, 28, J» 8.1 Hz), 0.91 (t, • 2H, J = 8.4 Hz), 3.60 (t, 2H, J = 8.4 Hz), 3.61 (t, 2H, J * , 8.1 Hz), 5.80 (s, 2H), 6.24 (s, 2H), 7.36 (m, 2H), 7.47 (dd, 1H, J = 9.0, 2.1 Hz), 7.57 (d, ÍH, J = 9.0 Hz ), 7.62 (m, ÍH), 7.91 (m, ÍH), 8.73 (d, ÍH, J = 2.1 Hz). Analysis (C26H37ClN402Si2) C, H, Cl, N. (e) Intermediate 3e-5-Phenyl-1 - [2- (trimethylsilanyl) -ethoxymethyl] -3-. { 1- [2- (trimethylsilanyl) * ethoxymethyl] -lH-benzoimidazol-2-yl} -lff-indazole: A mixture of 3d (192.0 mg, 0.363 mmol), phenylboronic acid (66.4 mg, 0.544 mmol), palladium (II) acetate (3.3 mg, 0.0145 mmol), CyMAP-1 (See Oíd et al. J. Am, Chem. Soc., 120, 9722 (1998) with a similar procedure) (3.7 g, 0.0145 mmol), and cesium fluoride (165 mg, 1.09 mmol) in, 1,4-dioxane (3.6 ml) it was heated in an oil bath to 100 ° C for 1 hour. After cooling to room temperature, the mixture was diluted with ethyl acetate (20 ml) and filtered to remove the black precipitate. The filtrate was washed with aqueous 1M sodium hydroxide (20 ml), dried over magnesium sulfate, concentrated and purified by chromatography on silica gel (0 to 4% methanol in dichloromethane) to give 3e (107.0 mg, 52%). %) in slightly yellowish oil fort: Rf = 0.26 (dichloromethane); XH NMR (CDC13) d -0.15 (s, 9H), -0.04 (s, 9H), 0.86 (t, 2H, J = 8.1 Hz), 0.95 (t, 2H, J = 8.1 Hz), 3.61 (t, 2H, J = 8.1 Hz), 3.66 (t, 2H, J - 8.1 Hz), 5.85 (s, 2H), 6.28 (s, 2H), 7.37 (m, 3H), 7.49 (t, 2H, J = 7.5 Hz), 7.63- 7.80 (, 5H), 7.91 (m, ÍH), 8.88 (s, ÍH). Analysis (C32H42NO2Si2 * 0.4H2O) C, H, N.
Example 3-3- (1-Benzoimidazol-2-yl) -5-phenyl-1H-indazole: Tetrabutylammonium fluoride (1.0 M in THF, 3.16 mL) and 1,2-diaminoethane (95 mg, 1.58 mmol) were added. to intermediate 3e (90.2 mg, 0.158 mmol). The solution was heated in an oil bath at 70 ° C for 20 hours, then heated to reflux for a further 24 hours. After cooling to room temperature, the solution was diluted with ethyl acetate (30 ml) and washed with a saturated aqueous solution of sodium bicarbonate (20 ml). The organic layer was dried over magnesium sulfate, filtered, it was concentrated and purified by chromatography on silica gel (25 to 50% ethyl acetate in hexanes) to provide 3- (li? -Benzo? midazole-2?) -5-Fenyl-lii-Indazole 2 (33.9 mg, 69%) in the form of a white solid: Rf = 0.30 (50% ethyl acetate / hexanes); XH NMR (CDC13) d 7.21 * (quintet of d, 2H, J = 5.7, 1.5 Hz), 7.39 (t, ÍH, J - 7.4 Hz), 7.53 (t, 3H, J = 7.5 Hz), 7.76 (m , 5H), 8.71 (s, 1H), 13.01 (s, ÍH), 13.70 (s, 1H). HRMS calculated for C2oH? 5N4 311.1297 (MH +), found 311.1283.
Example 4: 3- [3- (lH-Benzoimidazol-2-yl) -lg-indazol-S-yl] -phenol [dioxane = dioxanoj (a > Intermediate 4a -5- (3-Methoxyphenyl) > ~ 1-. {2- (trimethylsilyl) -ethoxymethyl] -3- { 1- [2- (trimethylsilanyl) - ethoxymethyl] -lH-benzoimidazol-2-yl.}. -lfi-indazole: A mixture of the intermediate 3d (371.5 mg, 0.702 mmol), 3-methoxyphenylboronic acid (160 mg, 1.05 mmol), palladium acetate (II ) (7.9 mg, 0.0355 mmol), CyMAP-1 (See Oyd et al., J. Am. Chem. Soc., 120, 9722 (1998), "incorporated herein by reference, with a similar procedure) (14 mg, 0.0355 mmol), and cesium fluoride (320 mg, • 2.11 mmol) in 1,4-dioxane (7.1 ml) was heated in an oil bath at 90 ° C for 22 hours. After cooling until the } , 89 I. *. At room temperature, the mixture was diluted with ethyl acetate (50 ml) and filtered to remove the black precipitate. * The filtrate was dried over magnesium sulfate, concentrated; and purified by chromatography on silica gel (10% ethyl acetate in hexanes) to give 4a (178.3 mg »42%) as a slightly yellow oil: Rf = 0.20, (10% ethyl acetate / hexanes); XH NMR (CDC13) d -0.14 (s, 9H), -0.03 (s, 9H), 0.86 (t, 2H, J = 8.1 Hz), 0.95 (t, 2H, J- * »8.1 Hz), 3.61 ( t, 2H, J = 8.1 Hz), 3.66 (t, 2H, J »8.1 Hz), 3.91 (s, 3H), 5.85 (s, 2H), 6.27 (s, 2H), 6.93 (ddd, ÍH, J = 1.1, 2.5, 8.1 Hz), 7.27-7.40 (m, 5H), 7.63-7.70 5 (m,, 2H), 7.77 (d, IH, J = 1.5, 8.7 Hz), 7.93 (m, ÍH) 8.87 (e, ÍH). Analysis (C33H44N403Si2) C, H, N. (b) Example 4-3- [3- (l-Benzoimidazol-2-yl) -lH-indazol-5-yl] -phenol: A solution of intermediate substance 4a (88.3 mg, 0.147 mmol) in 1,2-dichloroethane (3.0 ml) was treated with a complex of boron-methyl sulfide trichromide (1.0 M in diechloromethane, 0.588 ml) and heated to reflux for 1 hour. Stirring was continued at room temperature for 16 hours. Water (5.0 ml) was added, and stirring continued for 30 minutes at room temperature. The mixture was diluted with diethyl ether (30 ml) and washed with saturated aqueous sodium bicarbonate solution (20 ml). The organic layer was washed with 1 M NaOH (3 x 30 ml). The , * & t 1 * í «l The aqueous wash combination was acidified to pH = 1 with 6 M HCl and extracted sequentially with ether (30 mL), ethyl acetate (30 mL), and dichloromethane (2 x 20 mL). The organic extracts were combined, dried over magnesium sulfate, filtered, concentrated and purified by chromatography on silica gel (50 to 75% ethyl acetate in hexanes) to give phenol 4 (11.6 g, 24%) in the form of white powder: lE NMR (DMSO-tíé) d 6.78 (dd, ÍH, J = 1.9, 7.7 Hz), 7.12-7.27 (m, 4H), 7.31 (t, 1H, J = 7.7 Hz), 7.52 (dd, ÍH, J = 2.1, 6.6 Hz), 7.71 (d, 2H, J = 1.1 Hz), 7.76 (dd, ÍH, J = 1.5, 6.8 Hz), 8.67 (s, 1H) , 9.57 (s, ÍH), 13.00 (s, ÍH), 13.68 (s, lH) HRMS calculated for 327. 1246 (MHT), found 327.1231.
Example 5: 3- (1-Benzoimidazol-2-yl) -5- (3-methoxyphenyl) -1H-mdazole (a) Example 5 -3- (lH-Benzoimidazol-2-yl) -5- (3-methoxyphenyl) -IH-indazole: The same crude reaction mixture from which example 4 was obtained also provided the ethoxyphenyl analogue 5 as follows: r ^ * í 91 A solution of intermediate 4a (88.3 tng, 0.147 mmol) in 1,2-ylichloroethane (3.0 ml) was treated with a complex of boron tribromide-methyl sulfide (1.0 H in dichloromethane, 0.588 ml) and heated to reflux for 1 hour. The stirring was then continued for 16 hours at room temperature. Water (5.0 ml) was added, and stirring continued for 30 minutes at room temperature. The mixture was diluted with diethyl ether (30 ml), and washed with a saturated aqueous solution of sodium bicarbonate (20 ml). The organic layer was washed with 1 M NaOH (3 x 30 ml). The organic layer was then dried, filtered, concentrated and purified by chromatography on silica gel (50 to 75% ethyl acetate and hexanes) to give 5 (9.3 mg, 19%) as a white powder: XE NMR (DMSO-d6) d 3.86 (s, 3H), 6.98 (dd, ÍH, J = 2.1, 7.8 Hz), 7.19-7.23 (m, 3H), 7.30 (d, ÍH, J = 7.8 HZ), 7.45 (t, 1H, J = 7.8 Hz), 7.52 (dd, ÍH, J = 1.8, 5.7 Ha), 7.75 (, 3H), 8.70 (s, ÍH), 13.00 (s, ÍH), 13.70 (s, lH ). HRMS calculated for C2? H16N4ONa 363.1222 (MNa +), found 363.1225.
¿A «a • 4- '' r 92 Indazol [dioxane = dioxane] (a) Intermediate 6a -5- (2-Fluorophen? l) -l- [2- (trimethylsilane) -ethoxymethyl] -3-. { l- [2- (Trimethylsilanyl) -ethoxymethyl] -lH-benzo? m? dazol-2-? l} -lH-? ndazol: A mixture of intermediate substance 3d (419.0 mg, 0.792 mmol), 2-fluorofen? lboromach acid (166 mg, 1.19 mmol), palladium (II) acetate (9.0 mg, 0.04 mmol), CyHUP- l (See Oíd et al., J. Am. Chem. Soc., 120, 9722 (1998) incorporated herein by reference, with a similar process) (16 mg, 0.04 mmol) and cesium fluoride (361 mg, 2.38 mmol ) in 1,4-d-oxane (8.0 ml) was heated in an oil bath at 70 ° C for 1 hour. When only a partial conversion was observed, more palladium (II) acetate (12 mg, 0.05 mmol) and CyMAP-1 (14 mg, 0.035 mmol) were added and stirring continued.
X: ~ '^' X '^' X- '"v.-Ü» 93 . 70 ° C for 16 hours. After cooling to room temperature, the mixture was diluted with ethyl acetate (50 ml) and filtered to remove the black precipitate. The filtrate was dried over magnesium sulfate, concentrated by evaporation. ",, and purified by chromatography on silica gel. (10% ethyl acetate in hexanes) to provide 6a (155.6 mg, 43%) as a colorless oil: XH NMR (CDC13) d - 0.1 (s, ': 9H), -0.03 (s, 9H), 0.86 (t, 2H, J = 8.1 HZ), 0.95 (t, 2H, < j ": - - = 8.1 Hz), 3.61 (t , 2H, J = 8.1 Hz), 3.66 (t, 2H, J = 8.1. 10 Hz), 5.86 (s, 2H), 6.27 (s, 2H), 7.15-7.39 (m, 5H), 7.57- .. 7.75 (m, 4H), 7.88 (m, iH) 8.82 (s, 1H), Analysis / '"' •;, (C32H4i N4? 2Si2 * 0.4H2O) C, H, N. (b) Example 6 3- (1H-Benzoimidazol-2-yl) -5- (2-fluorophenyl) -lH-indazole: 15'-r Example 6 was prepared by a method.
• Synthetic analogous to that of example 3. Treatment of intermediate substance 6a with tetrabutylammonium fluoride gave 6 (21.2 mg, 18%) as a color powder. "White: Rf = 0.35 (50% ethyl acetate / hexanes ); XH NMR; 20;; -'-. (DMSO-dβ) d 7.20 (m, 2H), 7.33-7.52 (m, 4H), 7.62- (m, 2H), 7.74 (m, 2H), 8.65 (s, ÍH), 13.02 (s, ÍH), 13.75 (s, lH) HRMS calculated for C20H14FN4 329.1202 (MH +) "found 329.1212 Analysis (C20H13FN4 • 1.1H20) C, H, N. *?: hí. *% 94 Example 7 ': 3- (1-Benzoimidazol-2-yl) -5- (4-methoxyphenylene) -1 indazole (a) Intermediate 7a '- 5-Iodo-3"- ((E) - 15 styryl) -1- (2-tr? met? ls? lanil-ethoxy? met l) -IH-indazole: The intermediate 7a 'was prepared with 5-nitromdazole (Acros organics, a division of Fisher Scientific, Pittsburgh, PA) in five steps according to the method used to prepare 6-Iodo-3- ((E) -styryl) -1- (2 - 0 trimethylsilanyl-ethoxymethyl) -lif-mdazole from 6-mtromdazole (Found in: Kania, Braganza, et al., Patent application "Compounds and Pharmaceutical Compositiones for Inhibitmg Protein Kmases, and Methods for Their Use", page 1 , 52, line 10 to page 53, line 26, and page 59, line 16 to page - 5 60, line 4, US Provisional Serial No. 60 / 142,130, i v 4 95 filed on July 2, 1999, incorporated herein by reference in its entirety): XH NMR (CDC13) d -0.06 (s, 9fej, 0.89 (t, 2H, J = 8.4 Hz), 3.57 (t, 2H, J = 8.4 Hz), 5.70 (S, 2H), 7.29-7.44 (m, 6H), 7.59 (d, 2H, J - 7.0 Hz), 7.67 (dd, ÍH, J = 8.7, 1.5 Hz), 8.36 (s, ÍH) (b) Intermediate 7b '- 5-Iodo-l- (2 * trimethylsilane-ethoxymethyl) -lH-indazole-3-carbaldehyde: Ozone bubbles were added to a solution of 5-iodo-3-styrene 2-SEM-mdazole 7a '(4.93 g, 10.35 mmol) in diechloromethane (500 ml) at -78 ° C. After 20 minutes the color of the solution changed from orange to deep blue.The mixture was purged with Argon for 30 minutes to remove the excess ozone, then dimethylsulfide (1.29 g, 20.7 mmol) was added, the cold bath was removed and the stirring continued for about 2 hours, until the internal temperature reached 15 ° C. washed with water (2 x 200 ml), dried over magnesium sulfate, filtered and concentrated. After chromatography on silica gel (10% ethyl acetate in hexanes) provided the aldehyde 7b '(2.74 g, 66%) as a yellow oil: XH NMR (CDC13) d -0.05 (s, 9H) , 0.89 (t, 2H, J = 8.4 Hz), 3.56 (t, 2H, J = 8.4 Hz), 5.79 (s, 2H), 7.43 (d, ÍH, J = 8.7 Hz), 7.76 (dd, ÍH, J = 8.8, 1.5 Hz), 8.71 (s, 1H), 10.22 (s, ÍH). ^ f (c) Intermediate substance 7 < 2'-3- (lH-Benzo? M? Dazol-2-? L) -5-iodo-l- (2-tr? Methyl? Lanyl-ethoxymethyl) -lH-mdazole: > To a solution of aldehyde 7b '(2.74 g, 6.81 irniol.ff in DMF (130 ml) was added 1,2-phenolylenediamine (0.74 g, 6.01 mmol) and elemental sulfur (0.26 g, 8.2 mmol). The mixture was heated in an oil bath at 95 ° C for 14.5 hours / cooled to room temperature, and diluted with ethyl acetate (500 ml) The solution was washed with a mixture of aqueous sodium chloride saturated (100 ml) and water f (100 ml) The organic layer was then washed with saturated aqueous sodium licarbonate (100 ml), followed by water (100 ml), dried over magnesium sulfate, filtered, The mixture was concentrated and purified by chromatography on silica gel (20% ethyl acetate in hexanes) to give impure 7q 'as a pale yellow solid, and precipitation in chloroform / hexanes gave pure 7c' (2.15 g, 64%). %) in the form of white powder: Rf = 0.23 (20% ethyl acetate / hexanes); XH NMR (DMS0-d6) d -0.12 (s, 0), 0.82 (t, 2H, J = 7.9 Hz), 3.59 (t, 2H, J = 7.9 Hz), 5.87 (s, 2H), 7.23 (m, 2H), 7.52 (d, 1H, J = 7.2 Hz), 7.73- 7.84 (m, 3H), 8.94 (s, ÍH), 13.13 (s, 1H). HRMS calculated for C2oH23IN4Os? 491.0759 (MH +), found 491.0738.
Y* (d) Intermediate 7d'-3- < lH- -Benzo? p? idazol-2-yl) -5- (4-methoxyphenyl) -1- (2-trimethylsilaftiJ.- ethoxymethyl) -lfi-indazole: A 2M aqueous sodium carbonate solution (6.4 ml) was added to a solution of 7c '(2.50 g, 5.10 mmol), 4-methoxy phenylboronic acid (1.01 g, 6.63 mmol) and tetrakis (triphenylphosphmo) palladium (0.59 g, 0.51 mmol) in 1,4-dioxane (35 ml) and methanol (15 ml). The mixture was heated to reflux for 5 hours, then cooled and partitioned between ethyl acetate (300 ml) and a mixture of saturated aqueous sodium chloride (100 ml) and water (100 ml). The organic layer was dried over magnesium sulfate, filtered, concentrated and purified by chromatography on silica gel (20% ethyl acetate in hexanes) to give a dark brown solid. The precipitation in dichloromethane / hexanes gave pure 7d '(948.6 mg, 40%) as a white powder: f = 0.13 (20% ethyl acetate / hexanes); XH NMR (DMSO-d6) d -0.10 (s, 9H), 0.85 (t, 2H, J = 7.9 Hz), 3.63 (t, 2H, J = 7.9 Hz), 3.82 (s, 3H), 5.91 (s) , 2H), 7.10 (d, 2H, J = 8.7 Hz), 7.23 (m, 2H), 7.43 (m, 1H), 7.54 (d, ÍH, J - 6.8 Hz), 7.69 (d, 2H, J = 8.7 Hz), 7.80 (m, ÍH), 7.92 (d, ÍH, J = 8.9 Hz), 8.70 (s, ÍH), 13.08 (s, ÍH). (e) Example 7'-3- < lH-Benzoimidazol-2-yl) -5- < 4metoXi enyl) -lfí-indazol: A solution of intermediate 7d '(148.4 mg, 0.315 mmol) in ethyl acetate (15 ml) at -78 ° C was treated with boron tribromide (1.0 M in dichloromethane, 4.73 ml). The solution was stirred for 17 hours, allowing the mixture to gradually warm to reach room temperature. Water (10 ml) was added, and the mixture was stirred at room temperature for 6 days. The solution was treated with a 3 M sodium hydroxide solution to a pH of 10, then extracted with ethyl acetate (3 x 20 ml). The combined organic extracts were dried over magnesium sulfate, filtered and concentrated. Purification by chromatography on silica gel (50% ethyl acetate in hexanes) provided 7 '(60.5 mg, 56%) gn as a white solid: Rf = 0.21 (50% ethyl acetate / hexanes); 1H NMR (DMSO-d6) d 3.82 (s, 3H), 7.08 (d, 2H, J = 8.9 Hz), 7.21 (m, 2H), 7.53-7.78 (m, 6H), 8.66 (s, ÍH), 12.96 (s, ÍH), 13.63 (s, ÍH). Analysis (C2? H? 6N4O-0.25CH2C12) C, H, N. 2i • Example 8 ': - [3- (lH-Benzo? M3.dazol-2 »-? L) -lH- ^ ndazol-5? L] -phenol xfa > A mixture of 7 'anisole (44.6 mg, 0.131 mmol) and pipdin hydrochloride (912 mg, 7.9 mmol) was heated in an oil bath at 180 ° C for 3 hours. Pindma salt 0 is liquid at this temperature. After cooling to room temperatureThe mixture was partitioned between ethyl acetate (20 ml) and saturated aqueous sodium bicarbonate (15 ml). The aqueous layer was then extracted further with ethyl acetate (3 x 20 ml). The combined organic extracts were dried over magnesium sulfate, filtered, concentrated and purified by chromatography on silica gel (50% ethyl acetate / hexanes) to give pure phenol 8 '(29.4 mg, 69%) as of a pale yellow solid: Rf = 0 0.23 (60% ethyl acetate / hexanes); XH NMR (DMSO-ds) d 6.91"(d, 2H, J = 8.4 Hz), 7.21 (m, 2H), 7.53 (m, 3H), 7.68 (s, 2H), 7.75 (d, ÍH, J = 6.9 Hz), 8.61 (s, 1H), 9.53 s, ÍH), 12.98 (s, ÍH), 13.63 (s, 1H) HRMS calculated for C20H14N4O 327.1256 (MH +), found 327.1253 Analysis (C20H? 3N4O »0.8 5 DMSO) C, H, N. * < * - «, * ? , r%. * t phenyl) -lj-T-indazole (a) Intermediate 9a'- 3-Methoxy-2-metuphenylamin: A suspension of 2-met? i-3-n? troanisole (Aldrich Chemicals) (8.87 g, 53 mmol) and palladium on 10% carbon (800 mg) in ethanol (400 ml) was stirred at hydrogen or 40 psi for 1 hour. After being filtered through a pad of Celite, the solution was concentrated and purified by chromatography on silica gel (30% ethyl acetate in hexanes) to provide aniline 9a '(6.94 g, 95%) as an oil slightly orange in color: f = 0. 20 (25% ethyl acetate / hexanes); XW WÍW (DMSO-d) d 1,88 (s, 3H), 3.68 (s, 3H), 4.74 (br s, 2H), 6.17 (d, ÍH, J = 8.1 Hz), 6.26 (d, ÍH, J = 8.1 Hz), 6.81 (t, ÍH, J = 8.1 Ht) * Analysis (C8HuNO) C, H, N. (b) Intermediate 9b'-l-Iodo-3-methoxy-2-methyl-benzene: 3-Methoxy? -2-met? L-fen? Lamma was diazotized (5.28 g, 38.5 mmol) according to the method of DeGra, et al. [DeGra,,, J.I .; Brown, V.H .; Colwell, .T .; Morpson, NE, J. Med. Chem., 17, 762 (1974)], incorporated herein by reference, which yielded aryl iodide 9b '(4.17 g, 44%) as a yellow oil: Rf = 0.53 (10% ethyl acetate / hexanes); XH NMR (CDC1) d 2.36 (s, 3H), 3.80 (s, 3H), 6.81 tm, 2H), 7.42 (dd, ÍH, J = 7.5, 1.5 Hz). (c) Intermediate substance 9c'- 2- < 3-Methoxy-2-pt txl-phenyl) -4,4,5,5-tetramet? L- [1,3,2] dioxaborolan: l-Iodo-3-methox? -2-met l-benzene was dissolved (3.80 g, 15.3 mmol), Bis (pmacolato) diborro (4.28 g, 16.8 mmol), potassium acetate (4.51 g, 46.0 mmol), and 1,1'-bis (diphenylphosphmo) ferrocene dichloropalladium (II) (625 mg, 0.766 mmol) in DMSO (70 ml) and heated to 80 ° C internal temperature for 1 hour. After cooling, the mixture was diluted with toluene (400 ml), washed with water (2 x 100 ml), dried over magnesium sulfate, filtered and concentrated. Purification by gel chromatography - rm t t silica (5 to 20% ethyl acetate in hexanes) provided "boronic ester 9c '(19.6 g, 52%) as a white crystalline solid: Rf = 0.27 (5% ethyl acetate / hexanes); AH NMR ( CDC13) d 1.34 (s, 12H), 2.42 (s, 3H), 3.81 (s, 3fi) «5 € .91 (d, 1H, J = 8.1 Hz), 7.14 (t, ÍH, J = 7.8 Hz) , 7.34 (d, * *,? ~. ÍH, J - 7.5 Hz) Analysis (C? 4H2? B03) C, H. (d) Intermediate 9d'-3- (lH-Benzoimidazol-2-yl) -5- (3-methoxy-2-methyl-phenyl) -1- (2-trimethylsilanyl-ethoxymethyl) -lff-indazole: 0 A solution of aqueous sodium carbonate (2M, 2.65 ml) was added to a solution of 7c. '(519.4 mg, 1.06 mmol), boronic ester 9c' (262.8 mg, 1.06 mmol), and 1,1'-bis (diphenylphosphmo) ferrocene dichloropalladium (II) (43.2 mg, 0.053 mmol) in DMF (12 ml). The mixture was heated in an oil bath at 75 ° C for 4.5 hours, then cooled and partitioned between ethyl acetate (100 ml) and a mixture of saturated aqueous sodium chloride (50 ml) and water (50 ml). The organic layer nica dried over magnesium sulfate, filtered, concentrated, but 1H NMR of this crude material showed only 60% 0 conversion. The crude mixture was redissolved in DMF (12 ml) and additional boronic ester * (253 mg, 1.01 mmol), catalyst (140 mg, 0.17 mmol) and sodium carbonate solution (2.65 ml) were added. Stirring continued at 80 ° C for 15.5 hours. After the same process as above, XH NMR crude showed less than 5% of 7c 'remnant. Purification by chromatography on f fc * - - *; of silica (10 to 30% ethyl acetate in hexanes) gives 9 d '(410.7 mg, 80%) as a white foam * Rf = 0.37 (30% ethyl acetate / hexanes, equal to 7c'); XH MÉJ (DMSO-de) d -0.10 (s, 9H), 0.85 (t, 2H, J = 7.9 Hz), 2.06 (s, 3H), 3.64 (t, 2H, J = 7.9 Hz), 3.85 (s) , 3H), 5.92 (s, 2H), 6.92 (d, 1H, J = 7.2 Hz), 7.02 (d, 1H, J = 8.3 Hz), 7.17-7.30 (, 3H), 7.47-7.53 (m, 2H ), 7.70 (d, 1H, J = 7.7 Hz), 7.90 (d, 1H, J = 8.7 Hz), 8.45 (s, ÍH), 13.09 (s, 1HH-Analysis (C28H32N? 2SY O. 3H2?) C, H, N. (e) Example 9'-3- (1H-Benzoimidazol-2-yl) -5- (3-methoxy-2-methyl-phenyl) -IH-indazole: In a manner analogous to example 3 , the treatment of intermediate 9d * with fluoride and tetrabutylamome gave 9 '(47.2 mg, 30%) as white powder: Rf = 0.23 (5% methanol / dichloromethane); XH NMR (DMSO-de) d 2.07 (s, 3H), 3.85 (s, 3H), 6.91 (d, ÍH, J = 7.4 Hz), 7.01 (d, 1H, J = 8.1 Hz), 7.24 (m, 3H), 7.39 '(d4 / 1H, J = 8.7, 1.5 Hz), 7.50 (m, ÍH), 7.68 (d, 2H, J - 8.5 Hz), 8.40 (s, ÍH), 12.96 (s, ÍH) , 13.66 (s, ÍH). Analysis (C22H? 8N4O'0.3H2O) C, H, N.
? Example 10 ': 3- [3- (lH-Benzoimidazol-2-j.l) -lii-indazol-5 »l]'? Xr¡ Phenol 10 'was prepared by a synthetic method analogous to that of phenol 8', by treatment of 9 * (31.6 mg, 0.089 mmol) with pyridine hydrochloride provided 10 'phenol (20.8 mg, 70%) in the form of a solid whitish in color: Rf = 0.21 (60% ethyl acetate / hexanes); "H NMR (DMS0-d6) d 2.04 (s, 3H), 6.75 (d, ÍH, J - 7.0 Hz), 6.85 (d, ÍH, J = 7.7 Hz), 7.08 (t, 1H, J = 7.7 Hz ), 7.19 (qumt, 2H, J = 7.7 Hz), 7.39 (dd, 1H, J = 8.7, 1.5 Hz), 7.50 (d, ÍH J = 7.5 Hz), 7.68 (m, 2H), 8.39 (s, ÍH), 9.39 (s, ÍH), 12.95 (s, 1H), 13.64 (s, 1H) HRMS calculated for C2? H? 6N40 341.1402 (MH "), found 341.1410. Analysis (C2? H? 6N4O'1.0MeOH) C, H, N.
Example 11: 5- (2-Methylphenol) -3-phen l-lH-indazole • \ »'JT * 105 I OH 11e 11 [Toluene = Toluene, Dioxane = Dioxane] (a) Intermediate substance lla - 5-N? Tro-3-phenyl-1-mdazole: To a solution of 2-chloro-5-n? Trobenzophenone (15 g, 57 mmol) in ethanol (300 ml) was added hydracma monohydrate (50 ml, 1 mol). The resulting solution was stirred overnight (16 hours) at room temperature, then poured into water (2 1) and stirred for another 2 hours. The precipitate that formed was collected by filtration, washed with water (2 x 100 ml), and dried with air to provide 5-N? Tro-3-phenol-lH-mdazole (13.1 g, 80%) in the form of a yellow solid: XH NMR (DMSO-d6) d 7.48 (tt, 1H, J = 1.3 , 7.4, Hz), 7.58 (dd, 2H, J = 7.1, 7.4 Hz), 7.78. { $, 1H, J = 9.2 Hz), 8.01 (dd, 2H, J = 1.3, 7.1 Hz), 8.25 (dd, "ÍH, J = 2.1, 9.2 Hz), 8.91 (d, ÍH, J = 2.1 Hz) 13.88 (s 1H) Analysis (C? 3H9N302) C, H, N. (b) Intermediate llb-5-N? Tro-3-phen? II- [2- (trimethylsilanyl) ethoxymethyl] lH-mdazole: Dnsopropylethylamine (15 ml, 86.1 mmol) was added dropwise to a solution of 5-n? Tro-3-phen? Ll # -indazole (13 g, 54.3 mmol) and 2- (trimethylsilyl) ethoxymethyl chloride (15 g. 90 mmol) in acetonitrile (400 ml) The resulting reaction mixture was stirred at room temperature for 2 hours, then poured into water (11) and extracted with ethyl acetate (3 x 300 ml). The combined organic extracts were dried over sodium sulfate and concentrated, the residue obtained was dissolved in toluene (40 ml), and 2- (trimethylsilyl) ethoxymethyl chloride (3 ml, 17 mmol), tetrabutylammonium bromide (500 ml) was added to this solution. mg) and silica (40 g) This mixture was stirred overnight at room temperature and Then it leaked. Subsequently the filtrate was concentrated. Chromatography was performed on silica gel (5% ethyl acetate / hexanes), which yielded llb (15 g, 75%) in the form of a yellow solid: XH NMR (DMSO-de) d - 0. 11 (s, 9 H) 0.83 (t, 2H, J = 7.9 Hz), 3.62 (t, 2H, J = * 7.9 Hz), 5.91 (s, 2H), 7.52 (tt, ÍH, J = 0.7, 7.4 Hz), 7.60 (dd, • 2H, J = 7.1, 7.4 Hz), 8.00 (d, ÍH, J = 9.2 Hz), 8.02 (dd, 2H, J = 0.7, 7.1 Hz), 8.35 (dd, ÍH, J = 2.1, 9.2Hz), 8.91 (d, 1 ?, J = 2.1 Hz). (c) Intermediate 11c-5-Amino-3-phenyl-1- [2- (tnmet? lysian? l) ethoxy? met? l] -lH-? ndazole: A mixture of S-nitro-S-phenyl- l-1 [2- (trimethylsilanyl) ethoxymethyl] lH-mdazole lb (14 g, 37.9 mmol) and 10% palladium on carbon (1 g) in ethyl acetate (500 ml) was stirred under an atmosphere of hydrogen during the night. The reaction mixture was filtered through celite, then concentrated to provide 11c (12.2 g, 95%) in the form of a white solid: XH NMR (DMSO-d6) d -QJ12 (s, 9 H) , 0.80 (t, 2H, J = 8.0 Hz), 3.54 (t, 2H, J = *, i8.0 -Hz), 5.01 (br s, 2H), 5.67 (s, 2H), 6.89 (dd, ÍH, J - 1.8, 8. 8 Hz), 7.12 (d, ÍH, J = 1.8, Hz), 7.37 (tt, 1H, J = 0.5, 7. 4 Hz), 7.47 (d, ÍH, J = 8.8 Hz), 7.50 (dd, 2H, J = 7.2, 7. 4 Hz), 7.87 (dd, 2H, J = 0.5, 7.2 Hz). (d) Intermediate substance lid-5-i © do-3-phenyl-l- [2- (tr? methylsilaml) ethoxy? met? l] -lH-mdazole: Intermediate He was dissolved (12 g, 35.3 mmol) in a mixture of acetic acid (300 ml) and water (50 ml). The mixture was cooled in an ice bath and salt at a temperature of -5 ° C. A mixture was slowly added * - & 108 solution of sodium nitrite (4.5 g, 65.2 mmol) in water, (10 ml) at a rate such that the reaction temperature could be maintained below 3 ° C. The resulting diazonium solution was stirred at a temperature of 0 ° C for 20 minutes. A solution of potassium iodide (6.5 g, 39.2 mmol) in water (10 ml) was slowly added to the reaction, again at such a rate that the reaction remained below 3 ° C. Then the reaction was allowed to stir overnight, gradually equilibrated until reaching t at room temperature. The crude reaction mixture was poured into water (300 ml) and extracted with ethyl acetate (2 x 500 ml). The combined organic extracts were dried over sodium sulfate and concentrated. Chromatography was performed on silica gel (5% ethyl acetate / hexanes), which afforded lid (4 g, 25%) in the form of a yellow oil: aH NMR (DMSO-de) d -0.12 (s) , 9 H), 0.83 (t, 2H, J = 7.9 Hz), 3.57 (t, 2H, J * 7.9 Hz), 5.80 (s, 2H), 7.45 (tt, ÍH, J = 1.3, 7.5 Hz), 7.54- (dd, 2H, J »7.1, 7.5 Hz), 7.67 (d, ÍH, J = 8.8 Hz), 7.75 (dd, ÍH, J = 1.5, 8.8 Hz), 7.94 (dd, 2H, J = 1.3 , 7.1 Hz), 8.40 (d, ÍH, J = 1.5 Hz). (e) Intermediate substance He-5 - (^ - Methylfeml) -3-pheny1-1- [2 - (trimethylsilanyl) ethoxymethyl] -lphi-indazole: Aqueous saturated sodium bicarbonate (2 ml) was added to a mixture of the intermediate lid (130 mg 0.3 mmol), 2-methylphenylboronic acid (120 mg, 0.9 mmol) and tetrakis (triphenylphosphine) palladium (0) (25 mg, 0.02 mmol) in 1,4-dioxane (10 ml). The resulting reaction mixture was heated in an oil bath at a temperature of 90 ° C for 18 hours. After cooling to room temperature, the crude reaction mixture was poured into water (50 ml) and extracted with ethyl acetate (2 x 25 ml). The combined organic extracts were dried over sodium sulfate and concentrated. Chromatography was carried out on Cfel * (silica (10% ethyl acetate / hexanes), which yielded He (100 mg, 84%) in the form of a whitish solid: XH NMR (DMSO-de) d - 0.10 (s, 9 H), 0.85 (t, 2H, J - 8.0 Hz), 2.24 (s, 3H), 3.62 (t, 2H, J - 8.0 Hz), 5.85 (s, 2H), 7.29 (m , 4H), 7.42 (tt, ÍH, J = 1.4, 7.4 Hz), 7.47 (dd, ÍH, = 1.5, 8.3 Hz), 7.52 (dd, 2H, J = 7.1, 7.4 Hz), 7.84 (d, ÍH , J = 8.3 Hz), 7.93 (d, 1H, J = 1.5 Hz), 7.99 (dd, 2H, J = 1.4, 7.1 Hz). (F) Example 11-5- (2-Methylphenyl) -3-phenyl -lH-indazole: Tetrabutylammonium fluoride (1.0 M in THF, 2 ml) was added to a solution of the intermediate He (100 mg, 0.24 mmol) in tetrahydrofuran (5 ml) This solution was heated in an oil bath at a temperature X 110 60 ° C for 18 hours, then poured into water (25 ml) and extracted with ethyl acetate (2 x 25 ml). The extracts.
The combined organics were dried over sodium sulfate and concentrated. Chromatography was performed on silica gel (20% ethyl acetate / hexanes), which gave 5- (2-methylphenyl) -3-phenyl-1H-indazole 11: (55 mg, 80%) in the form of a solid. whitish color: XH NMR (DMSO-de) d 2.24 (s, 3H), 7.28 (m, 4H), 7.37 (dd, 1H, J = 1.5, 8.6 Hz), 7.38 (tt, ÍH, J = 1.4, 7.5 Hz), 7.50 (dd, 2H, J = 7.1, 7.5 Hz), 7.64 0 (d, ÍH, J = 8.6 Hz), 7.91 (d, ÍH, J = 1.5 Hz), 7.99 (dd, H, J = 1.4, 7.1 Hz), 13.30 (s, 1H). 7 Analysis (C20H16N2 »0.25HaO) C, H, N.
Example 12 3-Phenyl-5- [2- (trifluoromethyl) phenyl3-lH ~ 5 > . ". i I 111 (a) Intermediate 12a-3-Phenyl-5- [2 - * - (trifluoromethyl) phenyl] -1- [2-trimethylsilanyl) ethoxymethyl-1-indazole: The intermediate solution 12a was prepared through a synthetic method analogous to the intermediate substance. The palladium-catalyzed coupling of the intermediate substance lid with 2- (trifluoromethyl) phenylboronic acid resulted in 12a (48%) of a white solid. : XH NMR (DMSO-d6) d - 0.12 (S, 9H), 0.87 (t, 2H, J = 8.1 Hz), 3.72 (t, 2H, J = 8.1 Hz), 5.62 (s, 2H), 7.32 ( m, 1H) 7.38 (tt, ÍH, J = 0.8, 7, '* Hz), 7.48 (dd, 2H, J = 7.1, 7.4 Hz), 7.51 (m, ÍH), 7.63 (<; M, ~ ÍH, J = 7.2, 7.7 Hz), 7.66 (dd, ÍH, J = 1.6, 8.6 Hz), 7.75 (m, ÍH), 7.82 (d, ÍH, J = 8.6 Hz), 7.91 (d , ÍH, J = l.ß Hz), 7.96 (dd, 2H, J - 0.8, 7.1 Hz). (b) Example 12-3-Phenyl 5- [2- (trifluoromethyl) phenyl] -lH-indazole: The substance of Example 12 was worked up in a similar manner to Example 11. Treatment of 12a with tetrabutylammonium fluoride resulted in 3 -Fenyl 5- [2-trifluoromethyl) phenyl] -IH-indazole 12 (74%) as a solid * * white: 1H NMR (DMSO-d6) d 7.34 (m, 1H), 7.38 (tt, 1H, J * 1.3, 7.3 Hz), 7.49 (dd, 2H, J = 7.1, 7.3Hz), 7.52 (m, ÍH) «. 7.62 (dd, 1H, J = 7.4, 7.7 Hz), 7.65 (dd, ÍH, J = 1.9, 8.6 * Hz), 7.73 (dd, J = 7.2, 7.5 Hz), 7.85 (d, ÍH, J = 8.6 Hz * -,? R / * '»f - t 2. 22 (s, 3H), 3.62 (t, 2H, J = 8.0 Hz), 3.73 (t, 2H, J »8.1 Hz), 5.25 (s, 2H), 5.85 (s, 2H), 6.93 (dd, ÍH , J = 2.6, 8.3 Hz), 6.98 (d, ÍH, J = 2.6 Hz), 7.22 (d, ÍH, J = 8.3 Hz),? ! 7.43 (tt, 1H, J = 0.9, 7.7 Hz), 7.45 (dd, ÍH, J = 1.3, 8.6 Hz), 7.52 (dd, 2H, J = 7.2, 7.7 Hz), 7.82 (d / ÍH, J «8.6 Hz), 7.89 (d, ÍH, J = 1.3 Hz), 7.99 (dd, 2H, J = 0.9, > 7.2Hz). (b) Example 13-5- (4-Hydroxy-methyl-1-phenyl) -3-feml-lH-indazole: The substance of Example 13 was prepared in a manner similar to Example 11. Treatment of 13a with tetrabutylammonium fluoride gave 5- (4-hydroxy-2-methyl-lfene) -3- phenyl-lH-indazole 13 (75%) as a pale yellow solid: XH NMR (DMSO-d6) d 2.17 (s, 3 H), 6.66 (dd, ÍH, J = 2.3, 8.2Hz), 6.70 (d, ÍH, J = 2.3 Hz), 7.08 (d, ÍH, J = 8.2Hz), 7.32 (dd, 1H, J = 1.5, 8.6 Hz), 7.39 (tt, 1H, J = 1.4, 7.7 Hz), 7.50 (dd, 2H, J = 7.2, 7.7 Hz), 7.59 (d, 1H, J = 8.6 Hz), 7.83 (d, ÍH, J = 1.5 Hz), 7.97 (dd, 2H, J * 1.4, 7.2Hz) 9.28 (s, ÍH), 13.22 (s, ÍH). Analysis (C20 H16 'N2 O »0.8 H20) C, H, N.
Example 14: 3-Phenol-5- (pyrid-4-yl) -lH-indazole * 14 (a) Intermediate 14a-3-phenyl? -5- (p? R? D-4-yl) -1- [2-tr? Met? L?? L? L) ethoxymethyl] -lfl-indazole: The substance of Example 14a was prepared in a manner similar to Example He. The palladium-catalyzed coupling of the intermediate substance Hd with p? Pdm-4-boron acid co, gave substance 14a (76%) as a white solid: NMR (DMSO-d6) 6 - 0.11 (s, 9H), 0.84 (t, 2H, J = 7.9 Hz), 3.62 (t, 2H, J = 7.9 Hz), 5.86 (s, 2H), 7.46 (ct, ÍH, J = 1.1, 7.4 Hz ), 7.51 (d, ÍH, J = 8.3 Hz), 7.56 0 (dd, 2H, J = 7.1, 7.4 Hz), 7.80 (dd, ÍH, J = 1.4, 8.3 Hz), 7.85 (dd, 2H, J = 1.6, 4.5 Hz), 8.07 (day, 2H, J = 1.1, 7.1 * * Hz), 8.41 (d, ÍH, J = 1.4 Hz), 8.64 (dd, 2H, J = 1.6, 4.5 Hz).
Item 116 (a) Intermediate 14b-3 Phenyl-5- (pyrid-3-yl) -1- [2- (trimethylsilanyl) ethoxymethyl] -lff-indazole: Substance 14b 'was prepared in a manner similar to the intermediate substance He . The palladium-catalyzed coupling of the intermediate substance lid with pyridin-3-boronic acid gave substance 14b '(66%) as a white solid: XH NMR (DMSO-d6) d -0.10 (s) , 9H), 0.83 (t, 2H, J = 7.9 Hz), 3.63 (t, 2H, J * 7.9 Hz), 5.86 (s, 2H), 7.43 (tt, 1H, J = 1.2, 7.5 Hz), 7.51 (dd, ÍH, J = 4.7, 10 8.0 Hz), 7.54 (dd, 2H, J = 7.1, 7.5 Hz), 7.65 (d, ÍH, J = 8.6 Hz), 7.73 (dd, ÍH, J = 1.5, 8.6 Hz), 8.07 (dd, 2H, J = 1.2, 7.1 Hz), 8.18 (ddd, ÍH, J = 1.6, 2.3, 8.0 Hz), 8.32 (d, ÍH, J = 1.5 Hz), 8.56 (dd, 1H, J = 1.6, 4.7 Hz), 8.90 (d, ÍH, J = 2.3 Hz). 15, (b) Example 14b-3-Phenyl-5- (pyrid-3-yl) -l-indazole: Similarly to Example 11, treatment * e 14b 'with tetrabutylammonium fluoride provided 3-phenyl-5- (py d-3-yl) -lH-indazole 14b (79%) as a white solid: XH NMR (DMS0-d6) d 7.41 (tt, ÍH, J = 1.3, 7.4 Hz), 7.49 (dd) , ÍH, J = 4.7, 7.9 Hz), 7.53 (dd, 2H, J = 7.1, 7.4 Hz), 7.70 (d, 1H, J = 8.7 Hz), 7.76 (dd, ÍH, J = 1.5, 8.7 Hz) , 8.08 (dd, 2H, J = 1.3, 7.1 Hz), 8.17 (ddd, ÍH, J = 1.7, 2.0, 7.9 Hz), 8.31 (d, ÍH, J = 1.5 Hz) 8.56 (dd, ÍH, J = ai s 117 1. 7, 4.7 Hz), 8.99 (d, 1H, J = 2.0 Hz), 13.35 (s, Iph). Analysis (Cis H13 N3) C, H, N.
• / * Example 15: 2-Met? L-3- [3- ((E) -est? Ril) -lff-índagol-S-illfgrtol (a) Intermediate 15a '-5- (3-Methoxy-2-methyl-phenyl) -3- ((E) -styryl) -1- (2-tpmet ls? lan? l-ethoxymethyl) -lfi-indazole Intermediate 15a 'was prepared from substance 7a' (571.8 mg, 1.42 mmol) through a synthetic method which is analogous to 9d ', which provided an analogue of styryl 15a' (442.5 ramol, 66% ) in the form of a yellow oil: 2H NMR (DMSO-d6) d -0.10 (s, 9H), 0.83 (t, 2H, J = 8.1 Hz), 2.07 (s, 3 H), 3.58 (t, 2H, J -7.9 Hz), 3.84 (s, 3H), 5.79 (s, 2H), 6.91 (d, 1H, J = 7.6 Hz), 6.99 (d, ÍH, J = 8.3 Hz), 7.22-7.41 ( m, 5H), 7.56 (d, -2H, J = 5.1 Hz), 7.70-7.78 (m, 3 H), 8.09 (s, ÍH). (b) Intermediate 15b '-5- (3-Met? x? -2-methyl-phenyl) -3- ((E) -styryl) -lfl-mdazole: Substance 15b' was prepared in a manner similar to Example 3. Treatment of 15 a '(211.4 mg, 0.449 mmol) gave 15b' (132.7 mg, 87%) as a white foam: Rj = 0.38 (50% ethyl acetate / hexanes): XH NMR ( DMSO-de) 5 1.98 (s, 3 H), 3.84 (s, 3 H), 6.91 (d, ÍH, J = 7.5 Hz), 6.98 (d, ÍH, J = 8.1 Hz), 7.21-7.61 (m , 8H), 7.70 (d, 2H, J = 7.4 Hz), 8.05 (s, ÍH), 13.18 (s, ÍH). HRMS calculated for C 23 H 20 N 2 O 341.1648 (MH +), found 341.1638. Analysis (C23H2oN20 »0.2H20) C, H, N. (c) Example 15-2-Meth1-3-. { 3- ((E) -est? Nl) -1H-? Ndazol-5? LJ-phenol: The phenol 15 'was prepared in a manner similar to the phenol 8'. The treatment of intermediate 15b '(63.1 mg, 0.185 mmol) with pyridine hydrochloride afforded phenol 15' (39.7 mg, 66%) as a whitish solid: R = 0.24 (50% ethyl acetate / hexanes): AH NMR (DMSO-de) d 2.05 (s, 3H), 6.74 (d, 1H, J = 7.5 Hz), 6.83 (d, 1H, J = 7.9 Hz), 7.05 (t, ÍH, J = 7.7 Hz) , 7.25-7.62 (m, 7 H), 7.70 (d, 2H, J, '= 7.2Hz), 8.03 (s, ÍH), 9.34 (s, 1H), 13.16 (s, ÍH). HRMS calculated for C22H18N20 327.1497 (MH *), found 327.1487. Analysis (C22H? 8N2O »0.5H2O) C, H, N. of ethylhexanes (2: 1), s MgSO4 and concentrated. It is with hexanes-Et20 (in one 1. 09 g of 16a (55%). XH NMR (CDC13) d 8.51 (s, ÍH), 7.8 & (d, - '$ 1H, J = 8.4 Hz), 7.64 (d, ÍH, J = 7.2Hz), 7.58 (m, 2H), 7.48 (s, ÍH), 7.41 (m, 3H), 7.31 (m, ÍH), 3.59 (t, 2H, J = 7.3 Hz), 1.41 (s, 12H), 0.91 (t, 2H, J = 8.3 Hz), -0.06 (s, 9H). Analysis (C27H37N203SiB) C, H, N. (b) Intermediate 16b-4- [3- ((E) -Styryl) -l- (2-trimethylsilanyl-ethoxymethyl) -lH-indazol-5-yl] -isoquinoline : Intermediate 16a (0.218 g, 0.47 mmol), 4-bromoisoquinoline (0.082 g, 0.39 ramol) and Na2C03 (0.1 g, 0.95 mol) were combined with 3 ml of DME and 0.5 ml of water and the mixture was degassed and it was purged with argon. Tetrakis (triphenylphosphino) palladium (0) (0.023 g, 0.02 mmol) was added, the mixture was re-degassed and then heated to reflux under argon for 15 hours. Aqueous treatment as with intermediate 16a and chromatography on silica (hexanes-ethyl acetate in a ratio of 4: 1) yielded O.lβl (96%) e 16b: XH NMR (CDC13) d 8.59 ( s, ÍH), 8.13 (m, 2H), 7.97 (d, ÍH, J = 7.6 Hz), 7.73 (m, 3H), 7.58 (m, 3H), 7.50 (d, 2H, J = 9.5 Hz), 7.26 (m, 4H), 5.82 (s, 2H), 3.68 (t, 2H, J = 8.1 Hz), 0.97 (t, 2H, J = 8.3 Hz), -0.03 (s, 9H). Analysis (C3oH3? N3OSi »0.75 H20) C, H, N. (c) Example 16-4- [3- ((E) -Styryl) -lH-indazol-5-yl] -isoquinoline: A solution of 16b ( 0.17 g, 0.35 mmol) in 3.6 ml of IM tetrabutylammonium fluoride in THF and ethylenediamine (0.475 ul, 0.427 g, 7.1 mmol) was heated to the reflux point for 1 hour. The reaction was diluted with ethyl acetate and brought to pH 7 with 0.4 M HCl, washed with brine, dried over MgSO4, and concentrated. Chromatography on silica (hexanes: ethyl acetate in a ratio of 1: 1) gave 0.079 g. { 64%) of the substance 16 in the form of a white solid. 1H NMR (CDC13) § -10.20 (brs, 1H), 9.31 (s, ÍH), 8.59 (s, ÍH), 8.16 (s, 1H), 8.09 (d, ÍH, J = 7.2Hz), 7.93 (d , ÍH, J = 7.2Hz), 7.20-7.75, (m, 11H). Analysis (C24HpN3"0 .H20) C, H, N. > Example 17 4- [3 - ((E) -Estirili, ^ lH-indazol * S-l > | ft- quinoline and [Chloroquinoline = Chloroquinoline, Ethylenediamner * - Ethylenediamine, Re flux = Reflux] (a) Intermediate substance 17a - 4- [3- ((E) -Estyryl) - 1- (2-tpmethylsilanyl-ethoxymethyl) -lfi-indazole -5-yl] -quifioline: Substance 17a was prepared by a synthetic method analogous to intermediate 16b. With him ^. Using 4-cyoroquinolma, substance 17a was prepared with, a yield of 79%. 2H NMR (CDC13) d 8.99 (d, ÍH, J * 4.4 Hz), 8.21 (d, 1H, J = 7.9 Hz), 8.15 (s, ÍH), 7.95 (d, ÍH, J¡ = 8.4 Hz), 7.72 (m, 2H), 7.42-7.62 (m, 10H), 5.82 (s, 2H), 3.67 (t, 2H, J = 9.3 Hz), 0.97 (t, 2H, J = 8.3 Hz), -0.02 ( s, 9 H). Analysis (C3oH3? N3OSI »0.5 H20) C, H, N. (b) Example 17-4- [3- ((E) -Styryl) -lH-indazol-5-l] -quinoline: Example 17 was prepared in a manner similar to intermediate 16. Substance 17a was deprotigated to give 17 in a 50% level yield as a white solid. * H NMR (CDC13) 13.10 (brs, ÍH), 8.98 (d, ÍH, J = 4.4 Hz), 8.37 (s, 1H), 8.15 (d, 1H j = 8.4 Hz), 8.00 (d, ÍH, J = 8.4 Hz), 7.54-7.79 (m, 9H), 7.37 (m, 2H), 7.26 (m, ÍH). Analysis (C24H17N3 * 1.0 H2Q) C, H, N.
Example 18: 5- (4-P r d l) -3- (2-Pyrrol l) -IfHtnd zol 18a 18b 18c 18d OH (TVB OH n * y • K. 124 (a) Intermediate 18a-2-Fluoro-5-nitrobenzoyl chloride: A solution of 2-chloro-5-nitrobenzyl-oic acid (10.3 g, 56 mmol) in thionyl chloride (90 mL, 1.2 mol) was heated at reflux for 2 hours. The excess thionyl chloride was removed by concentration, under vacuum. The residue obtained was dissolved in ether (150 ml), then concentrated to give 2-fluoro-5-nitrobenzoyl chloride 18a (11.21 g, 99%) as an off-white solid, lH NMR (DMSO-de) d 7.62 (dd, ÍH, J = 9.1, 9.6 Hz), 8.48 (ddd, ÍH, J = 3.0, 6.9, 9.1 Hz), 8.60 (dd, 1H, J = 3.0, 6.3 Hz). Analysis (C7H3NO3CIF) C, H, N, Cl. (B) Intermediate 18b-1- (2 ~ Fluoro »* 5-nitrophenyl) -1- (1-pyrrol-2-yl) methanone: A solution of 2-Chloro-5-nitrobenzoyl-18a (10.04 g, 49 mmol) and pyrrole (3.4 mL, 3.29 g, 49 mmol) in 1,2-dichloroethane (110 mL) was cooled to a temperature of 0 ° C before A1C13 (6.61 g, 49.6 mmol) was added as a solid. The resulting reaction mixture was stirred overnight, and was gradually warmed to room temperature. Subsequently, the crude reaction was poured into a mixture of concentrated HCl (20 ml) and ice water (00 ml). After stirring for another 90 minutes, the layers were separated and the aqueous phase was extracted with CH2C12 (2 x 200 ml). The combined organic extracts were washed with water (200 ml) and saturated NaHCO 3 (200 ml), dried over sodium sulfate and concentrated. Chromatography was performed on silica gel (25% ethyl acetate / hexanes), which afforded 18b (7.23 g, 63%) in the form of a pale yellow solid: XH NMR (DMSOrde) d 6.28 (ddd, 1H). , J = 2.1, 2.3, 3.6 Hz), 6.74 (ddd, 1H, J = 1.3, 2.3, 2.5 Hz), 7.32 (ddd, ÍH, J = 1.3, 2.4, 3.6 Hz), 7.65 (dd, 1H, J = 9.0, 9.1 Hz), 8.39 (dd, ÍH, J = 3.0, 5.8 Hz), 8.45 (ddd, ÍH, J = 3.0, 4.4, 9.1 Hz), 12.33 (broad, ÍH). Analysis (CnH7N2O3F «0.1HC1) C, H, N. (c) Intermediate 18c-l- (2-Fluoror * -5- nitro eml) -1- (1- [2- (tpmethylsilanyl) ethoxymethyl] -Ifi- pitrol-2-yl) methanone: A solution of 1- (2-fluoro-5-n? trofen? l) -1- (1 H- p? rrol-2-? l) methanone 18b (1.72 g, 7.3 mmol) in THF (30 ml) was added dropwise, under an argon atmosphere, to a stirred suspension of NaH (350 mg, 8.75 mmol) in THF (15 ml) at 0 ° C. This mixture was stirred at a temperature of 0 ° C for 45 minutes before the addition of 2- (trimethylsilyl) ethoxymethyl chloride (1.70 g, 10.2 mmol) in a single portion as the pure liquid. The reaction mixture 50 ml). The combined organic extracts were washed with brine (60 ml), dried over sodium sulfate and concentrated. Chromatography was performed on silica gel (10% ethyl acetate / hexanes), which provided the "Substance 18c (2.24 g, 84%) in the form of a yellow syrup: XH NMR (DMSO-d6) d -0.07 (s, 9H) 0.83 (t, 2H, J = 7.8 Hz), 3.53 ( t, 2H, J = 7.8 Hz), 5.74 (s, 2H), 6.27 (dd, ÍH, J = 2.5, 4.0 Hz), 6.75 (dd, ÍH, J = 1.4, 4.0 Hz), 7.57 (dd, ÍH , J = 1.4, 2.5 Hz), 7.64 (dd, 1H, J = 9.0, 9.1 10 Hz), 8.29 (dd, ÍH, J = 3.0, 5.8 Hz), 8.45 (ddd, ÍH, J = 3.0, '4.6, 9.1 Hz). Analysis (C? 7H2? N204FSl) C, H, N. (d) Intermediate 18d-l- (5-Am? No-2-fluorophenyl) -1- (1- [2- (trimethylsilanyl) ethoxy? Met? l] -lH-pyrrole-2-? l) methanone: u A mixture of 1- (2-fluoro-5-n? trofen? l) -1- (1- [2- (trimethylsilanyl) ethoxymethyl] -lif- pyrrol-2?) methanone 18c (3.63 g, 10 mmol) and 10% palladium on carbon (365 mg) in ethyl acetate (90 ml) was stirred under a hydrogen atmosphere overnight. The reaction mixture was filtered through celite and then concentrated to provide 18d (3.30 g, 99%) as an amber colored syrup: aH NMR (DMSO-de) d -0.07 (s, 9 H) 0.82 (t, 2H, J = 8.0 Hz), 3.50 (t, 2H, J = 8.0 Hz), 5.12 (br s, 2H), 5.71 (s) , 2H), 6.20 (dd, 1H, J = 2.5, 3.9 Hz), 6.59 (dd, ÍH, J = 2.9, 5.6 HZ), 5 6.60 (dd, ÍH, J = 1.8, 3.9 Hz), 6.66 (ddd , ÍH, J = 2.9, 4.3, Í 4 £? 127 8. 8 Hz), 6.93 (dd, ÍH, J = 8.8, 9.7 Hz), 7.42 (dd, ÍH, J »1.8, 2.5 Hz). Analysis (C? 7H23N202FSI) C, H, N. (e) Intermediate 18e-1- (2-Flaor-5-iodophenyl) -1- (1- [2- (trimethylsilanyl) ethoxymethyl] -lfi-pi -2-) methanone: Intermediate 18d (332 mg, 1.0 mmol) was dissolved in a mixture of acetic acid (10 ml) and acetonitrile (10 ml). The solution was stirred vigorously and then cooled in an ice-bath and salt to a temperature of -5 ° C, before adding a solution of sodium nitrite (83 mg, 1.2 mmol) in water (10 ml). The resulting diazonium solution was stirred for 45 minutes and gradually warmed to 5 ° C. The reaction was again cooled to a temperature of -5 ° C prior to the addition of a solution of potassium iodide (232 mg, 1.4 mmol) in water (3 ral). The resulting mixture was stirred for another 2 hours, with heating to reach 15 ° C, then poured into a mixture of K2CO3 (30 g) and ice water (100 ml). This aqueous mixture was extracted with ethyl acetate (2 x 50 ral). The combined organic extracts were washed with 10% aqueous NaS03 (50 mL), dried over sodium sulfate and concentrated. Chromatography was performed on silica gel (5% ethyl acetate / hexanes), which afforded substance 18e (160 mg, 36%) in the form of a colorless oil: H NMR (DMSO-de) d -0.08 (s) , 9H), 0.81 (t, 2H, J = 7. 9 Hz), 3.50 (t, 2H, J = 7.9 Hz), 5.71 (s, 2H), 6.24 (dd, ÍH, J = 2.6, 4.0 Hz), 6.63 (dd, ÍH, J = 1.7, 4.0 Hz) , 7.18 • - (dd, 1H, J = 8.7, 9.7 Hz), 7.51 (dd, ÍH, J = 1.7, 2.6 Hz), *, s X 7.74 (dd, ÍH, J = 2.3, 6.4 Hz), 7.90 (ddd, ÍH , J = 2.3, 4.9,, * 8.7 Hz). Analysis (C? 7H2? N02FSÍI) C, H, N, I. (f) Intermediate 18f-l- [2-Fluoro-5- (4-pyridyl) phenyl] -1- (1- [2-trimethylsilanil) ethoxymethyl] -1J ~ -, * pyrrol-2-yl) methanone: Diisopropylethylamine (1.3 ml, 7.5 mmol) was added to a mixture of 1- (2-fluoro-5-iodophenyl) -1- (1- [2 - tpmethylsilanyl) ethoxymethyl] -lif-pyrrol-2-yl) methanone 18e (798, * mg, 1.8 mmol), tetrakis (triphenylphosphma) palladium (0) (65 mg, 0.06 mmol) and pyridin-4-boronic acid (323 mg, 2.6 mmol) in DMF (20 ml). The resulting reaction mixture was heated in an oil bath at a temperature of 90 ° C for 18 hours under an argon atmosphere. After cooling to room temperature, the crude reaction mixture was poured into water (100 ml) and extracted with ethyl acetate (2 x 75 ml). The combined organic extracts were washed with water (6 x 75 ml), dried over sodium sulfate and concentrated. Chromatography was performed on silica gel (20% ethylacetate / CH2Cl2), which gave the "substance 18f (407 mg, 57%) in the form of a pale yellow oil: XH NMR (DMSO-d6 ) d -0.06 (s, 9H) 0.84 (t, 2H, J = 7.9 Hz), 3.54 (t, 2H, J = 7.9 Hz), 5.76 (s, 2H), J *, 6. 24 (dd, 1H, J = 2.6, 4.0 Hz), 6.68 (dd, ÍH, J = 1.8, 4.0 Hz), 7.49 (dd, ÍH, J = 8.7, 9.3 Hz), 7.51 (dd, ÍH, J = 1.8, 2.6 Hz), 7.72 (d, 2H, J = 6.2 Hz), 7.87 (dd, ÍH, J = 2.4 ', 6.5 Hz), 8.02 (ddd, ÍH, J = 2.4, 4.9, 8.7 Hz), 8.63 (d, 2H, J = 6.2 Hz). Analysis (C22H25N202FSi) C, H, N. (g) Intermediate 18g-5- (4-Pyridyl) -3-Xl- [2- (trimethylsilanyl) ethoxymethyl] -lfi-pyrrol-2-yl) ? daz? > l: A solution of 1- [2-fluoro-5- (4-pyridyl) phenyl] -1- (1- [2-trimethylsilanyl) ethoxymethyl] -lif-pyrrol-2-yl) methanone 18f (504 mg, 1.3 mmol) and hydrazine monohydrate (1.7 ml, 35 mmol) in ethanol (35 ml) was heated to reflux for 42 hours. Then the ethanol was removed by concentration, under vacuum. The obtained residue was partitioned between water (25 ml) and ethyl acetate (25 ml). The layers were separated and the aqueous phase was extracted with ethyl acetate (25 ml). The combined organic extracts were washed with saturated NaHCO3 (30 ml), dried over sodium sulfate and concentrated. Chromatography was carried out on silica gel (3% CH0H / CH2C12), which gave substance 18g (430 mg, 87%) in the form of an off-white solid: XH NMR (DMSO-de) d -0.28 (s) , 9H), 0.63 (t, 2H, J = 8.0 Hz), 3.28 (t, 2H, J = 8.0 Hz), 5.72 (s, 2H), 6.26 (dd, ÍH, J = 2.8, 3.5 Hz), 6.79 (dd, ÍH, J = 1.7, 3.5 Hz), 7.10 (dd, 1H, J = 1.7, 2. 8 Hz), 7.67 (d, ÍH, J = 8.9), 7.77 (d, 2H, J = 6.2Hz), 7.81 (dd, ÍH, J = 1.6, 8.9 Hz), 8.19 (d, ÍH, J = 1.6 Hz), 8. 61 (d, 2H, J = 6.2Hz), 13.25 (s, "ÍH) Analysis (C:; H26N4OS?) C, H, N. (h) Example 18 - 5- (4-P? Ndal) - 3- (2-pyrrolidol) -lfl-indazole: Tetrabutylammonium fluoride (1.0 M in THF, 5 ml) was added to a solution of the intermediate 18g (366 mg, 0.9 mmol) and 1-2. d-ammoethane (150 mg, 2.5 mraol) in tetrahydrofuran (20 ml) This solution was refluxed for 42 hours, then poured into saturated NaHCO 3 (30 ml) * and extracted with ethyl acetate (2 x 25 ml). The combined organic extracts were dried over sodium sulfate and concentrated, chromatographed on silica gel (3% CH3OH / CH2Cl2), which afforded 5- (4- P? Pd? L) -3- (2 -p rrol? l) -líf-? ndazol 18 (71 mg, 29%) in the form of a whitish solid: XH NMR (DMSO-d6) d '6.20 (dd, ÍH, J = 2.6, 5.6 Hz ), 6.82-6.92 (m, 2H), 7.64 (d, 1H, J = 8.7 Hz), 7.81 (dd, ÍH, J = 1.4, 8.7 Hz), 7.83 (d, 2H, J = 6.1 Hz), 8.37 (d, 1H, J = 1.4 Hz), 8.62 (d, 2H, J * 6.1 Hz), 11.37 (s, ÍH), 13.09 (s, ÍH). Analysis (C? 6H12N4 * 0.05 CH2C12) C, H, N.
Example 18b ': 5-N? Tro-3- (2-P? Rrol l) -lfí-mdazol The substance 18b 'was prepared in a manner similar to the intermediate substance lla. The treatment of l- (2-flubro-5-nitrophenyl) -1- (1-pyrrol-2-yl) methanone 18b with hydrazine hydrate gave 5-nitro-3- (2-pyrrolyl) -lfi-indazole 18b ' (75%) in the form of a reddish-orange solid: XH JfR (DMSQ-d6) d 6.23 (ddd, ÍH, J = 2.4, 2.6, 3.6 Hz), 6.81 (d, ÍH, J = 1.5, 2.5, 3.6 Hz) 6.93 (ddd, 1H J = 1.5, 2.1, 2.6 Hz), 7.70 (d, ÍH, J = 9.2Hz), 8.21 (dd, ÍH, J = 2.0, 9 * J Hz), 8.90 (d, ÍH, J = 2.0 Hz), 11.57 (broad, ÍH), 13.62 (s, ÍH). Analysis (CnH8N402) C, H, N.
Example 19: 4- [3- (4-Chloro-lH-benzoimidaa Ql-2-yl) -lfi-indazpl »5-yl] -isoquinoline Zf, * rt * (a) Intermediate 19a-lfl-Inda2-l-3-carboxylic acid methoxy-m-amide: 0 3-Carboxydazole (100 g, 617 mmol) in 1-DMF was treated with carbonildumidazole (110 g, 678 mmol ) at a temperature of 25 ° C with evolution of gas during a lapse of 15 minutes. The reaction was heated to a temperature between 60 and 65 ° C for 2 hours and then cooled down to 25 ° C. N, O-dimethylhydroxylamine- 5 was added * Í, P "* -t 133 HCl (66.2 g, 678 mmol) in the form of a solid and the mixture was heated to 65 ° C for 3 hours. The reaction was concentrated to a paste and reduced in 21 C? FtCla, washed with water and then 2N HCl. It was observed that the product came out of the solution. The solid was filtered and rinsed separately with ethyl acetate. The ethyl acetate and CH2C12 layers were washed separately with NaHCO3 and brine, dried over MgSO4 and concentrated. The resulting solids were combined, triturated with a mixture of CH2C12 and ether in a ratio of 1: 1, filtered, dried to provide 106 g (84%) of intermediate 19a as a white solid: j = 0.38 (75% ethyl acetate in hexanes); 1H NMR (DMSO-dβ) d 13.60 (s, ÍH), 7.80 (d, ÍH, J = 8.2 Hz), 7.60 (d, ÍH, J = 8.2 Hz), 7.41 (t, 1H, J = 8.0 Hz) , 7.22 (t, ÍH, J = 8.0 Hz), 3.77 (s, 3H), 3.44 (s, 3H). Analysis (C10H11N3O2) C * H, N. (b) Intermediate 19b - 5-Iodo-lido-indazole-3-carboxylic acid co-methoxy-methyl-amide: To amide 19a (20 g, 97.4 mmol) in 1 1 CH2C12 added bis (trifluoroacetoxy) iodobenzene (46 g, 107 mmol) followed by adding in portions of iodine (14.84 g, 58.5 mmol) at a temperature of 25 ° C. After one hour, 600 ml of saturated Na2HS03 was added and a solid was started to precipitate which was filtered and rinsed with the surplus CH2C12 The filtrate was washed with brine, dried over MgSO4, concentrated and the remaining solid was triturated with a minimum amount of CH2C12. The combined solids were dried UP * • in vacuo over KOH to provide 29.26 g (91%) of 19b in the form of a pale white solid: R / = 0.31 (50% ethyl acetate in haxanes); XH NMR (DMSO-de) d 13.79 (s, ÍH), 8.39 (s, ÍH), 7.65 (d, ÍH, J - 8.7 Hz), 7.48 (d, ÍH, J = 8.7 Hz), 3.76 (s, 3H), 3.44 (s, 3H). Analysis (CÍOHÍSOIOZ) C, H. (c) Intermediate substance 19c - 5-Yodo-l- (4-methoxy-benzyl) -lfl-ndazol-3-carboxylic acid-co-methoxy-methyl-amide: To iodine 19b ( 15 g, 45.3 mraol) in 200 ml of THF was added in portions NaH (1.9 g of a dispersion of 60% mineral oil, 1.14 g, 47.6 mmol), with evolution of gas. After 15 minutes, the reaction was cooled to a temperature of 0 ° C and p-methoxybenzyl chloride (8.51 g, 54.4 mmol) was added followed by Nal (679 mg, 4.5 mmol). The mixture was heated to 45 ° C for 9 hours and allowed to cool to 25 ° C. The solution was diluted with ethyl acetate, washed with saturated aqueous NHC1, brine and dried over MgSO4 and concentrated to a viscous oil. Ether was added to the oil and a solid formed which was filtered and rinsed with ether to provide 14.18 g (70%) of 19c as a pale yellow solid: Rj = 0.42 (50% ethyl acetate in «*" - 135 * c 'habanos); XH NMR (CDC13) d 8.60 (s, 1H), 7.56 (dd, ÍH, J, "8.8, 1.6 Hz), 7.11 (m, 3H), 6.80 (dd, 2H, J = 6.7, 2.1 HzJ, 5.52 ( s, 2H), 3.81 (s, 3H), 3.75 (s, 3H), 3.51 (s, 3H). / Analysis (C? 8H18N302I) C, H, N, I. 5 (d) Intermediate substance 19d - 5 -Iodo-l- (-methoxy-benzyl) -lfi-indazole-3-carbaldehyde: Amide 19c (12.8 g, 28.3 mmol) in 300 ml of THF was cooled to -5 ° C and LiAlH was added in portions ( 1.29 g, 34 mmol) for 10 minutes After 1: after 30 minutes, the reaction was warmed by the slow addition of ethyl acetate at -5 ° C and poured into 0.4 N NaHS04.The organic layer was washed with water. brine, dried over MgSO4 and concentrated to give a slightly whitish solid which was triturated with a minimum amount of ether, filtered, washed with ether and dried to give 9.79 g (88%) of aldehyde 19d. as a white solid: Rf = 0.57 (50% ethyl acetate in hexanes), 1 H NMR (CDCl 3) d 10.20 (s, ÍH), 8.96 (s, ÍH), 7.63 (dd, 1 H, J = 8. 8, 1.6 Hz), 7.18 (m, 3H), 6.83 (d, ÍH, J = 8.7 Hz), 5.57 20 '' (s, 3H), 3. 75 (s, 3H). Analysis . (C? 6H? 3N2O2I »0. 1 ethyl acetate) C, H, N, I. (e) Intermediate 19e-l- (4-Mei? xibenzyl) -5- (4,4,5,5-tetramethyl- [1,3,2] -dioxaborolan-2-yl) -1H- Indazole -3-carbaldehyde: Bis (pmacolato) diboro (Aldrich Chemicals) (7.05 g, 27.8 mmol), iodide 19d (9.90 g, 25.24 mmol), potassium d® acetate (12.4 g, 126 mraol), and 1,1 '-bis (diphenylphosphine) ) ferrocene dichloropalladium (II) (515 mg, 0.631 mmol) was dissolved in dimethylsulfoxide (150 ml), degassed and heated in an oil bath at a temperature of 80 ° C for one hour. After cooling to room temperature, the mixture was partitioned between ethyl acetate (200 ml) and water (150 ml). The organic layer was dried over magnesium sulfate, filtered, concentrated and purified by chromatography on silica gel (25% ethyl acetate in hexanes) to give boronic ester 19e (9.75 g, 98%). 'as a whitish solid: Rf = 0.37 (25% ethyl acetate in hexanes); XH NMR (DMSO-de) d 1.31 (s, 12 H), 3.69 5 (s, 3 H), 5.75 (s, 2 H), 6.87 (d, 2 H. J - 8.7 Hz), 7.27 (d. 2 H, J = 8.7 Hz), 7.74 (d, 1H, J = 8.4 Hz), 7.91 (d, 1H, J = 8.4 Hz), 8.52 (s, ÍH), 10.17 (s, ÍH). Analysis. (C22H25BN20) C, H, N. (f) Intermediate substance 19f-5-Isoquinolin-4-yl-0 l- (4-methoxy-benzyl) -l-triazole-3-carbaldehyde To a degassed solution of boronic ester 19e (6.00 g, 15.30 mmol) and 4-bromoisoquinoline (5.17 g, 24.8 mmol) in ethylene glycol dimethyl ether (DME, 76 mL) was added. aqueous sodium carbonate solution (2.0 M, 38.2 ml 76.4 5 mmol) followed by tetrakis (triphenylphosphine) palladium (0) (883) stirred at room temperature for 10 minutes, filtered through a pad of celite, acidified with 3N aqueous hydrochloric acid, and extracted with dichloromethane (3 x 200 ml). The combined organic layers were dried over magnesium sulfate, filtered, concentrated and purified by chromatography on silica gel (1 to 20% ethyl acetate in dichloromethane), to give the diamine 19g (2.13 g, 60%) in form of a yellow oil: R / = 0.30 (dichloromethane); X H NMR (DMSO-d 6) d 4.60 (br s, 2 H), 4.80 (br s, 2 H), 6.37 (t, 1 H, J = 7.8 Hz), 6.48 (m, 2 H). Analysis (C6H7C1N) C, H, Cl, N. (h) Intermediate substance 19h-4- [3- (4-Chloro-lJi-r> benzo? M? Dazol-2-? L) -1- (4 -metox? -benc? l) -lfi-indazol-5-yl] - isoquinoline: The aldehyde 19f (405.6 mg, 1.03 mmol) and the diam 19g (147 mg, 1.03 mmol) were condensed in the presence of elemental sulfur (50 mg, 1.55 mmol) analogously to the synthesis of intermediate 7c ', which gave the intermediate substance 19h (275.5 mg, 52%) as a pale yellow solid R / = 0.12 (50% ethyl acetate in hexanes); XH NMR (DMSO-d6) d 3.74 (s, 3H). 5.83 (s, 2H), 6.93 (d, 2H, J = 8.8 Hz), 7.22 (m.2H), 7.38 (d, 2H, J = 8.5 Hz), 7. 48 (d, ÍH, J = 7.2 Hz), 7.67 (dd, 1 H, J = 8.7, 1.5 HE), 7. 76 (m, 3H), 8.04 (d, ÍH, J = 8.7 Hz), 8.26 (dd, 1 H, J = 7. 4, 1.5 Hz), 8.54 (s, ÍH), 8.64 (s, 1H), 9.40 (s, ÍH), 13.41 (s, ÍH). (i) Example 19-4- [3- (4-Chloro-1-benzo-m-dazol-2-yl) -lfl-ndazol-5-l] -α-soquol-nol: § Acid was added concentrated sulfuric acid (0.3 ml) to a solution of substance 19h (121.6 mg, 0.236 mmol) and tpfluoroacetic acid (3.0 ml), and stirred at room temperature for 19 hours. The mixture was then diluted coft water (50 ml), treated with concentrated aqueous ammonium hydroxide until a pH = 8 was reached and extracted with ethyl acetate (3 x 50 ml). The combined organic extracts were dried over magnesium sulfate, filtered, concentrated and purified by chromatography on silica gel to provide substance 19 (41.5 5 mg, 44%) as a white solid: Rf = 0.40 (75% ethyl acetate in hexanes), 1E NMR (DMSO-de) [Some peaks * are doubled due to tautomeric isomerization] d 7.22 (m, 2H), 7.48 (d, ÍH, J = 7.2 Hz), 7.64 (d, 1H, J = 8.7), 7.79 (m, 4H), 8.27 (d, IH, J = 7.5), 8.55 (S, IH), 8.63 (s, 1H), 9.40 (s, IH), 13.39 and 13.56 ( 2 s, ÍH as a whole), 13.94 (s, ÍH). Analysis, (C23H? 4ClN5 »l .2 CH3OH) C, H, Cl, N.
E xemployment 20: 4-. { 3- [5- (4-Met? L-p? Peyacm-l-? L) -lH-benzo? Pu.dazol-2-? 3 -lflundazol -5- l} -isoquin.pl ina After cooling to room temperature, the ethyl sol (150 ml) was washed and saturated aqueous sodium chloride (50 ml). The organic layer was dried over magnesium sulfate, filtered, concentrated and purified by chromatography on silica gel (1: 20: 400 conc. Aqueous NH 4 OH: EtOH: CH 2 C 12), which gave substance 20a (623 mg, 50%). %) as a brownish-orange colored foam Rf = 0.20 (10% ethanol in dichloromethane), XH AÚ «(DMSO-d6) [Some peaks double due to tautomeric isomerization] d 2.23 (s, 3 H), 2.49 (m, 4H), 3.10 (m, 4H), 3.71 (s, 3H), 5.80 (s, 2H), 6.91 (m, 4H), 7.36 and 7.47 (2 d, 3H as a whole, J = 8.3, 8.7 Hz), 7.64 (d, 1H, J = 8.9 Hz), 7.77 (m, 3H), 7.99 (m. ÍH), 8.25 (d, ÍH, J = 7.2 'Hz), 8.53 (s, ÍH), 8.62 and 8.64 (2 s, 1H as a whole), .3 '(s.1H). 12.78 and 12.83 (2s, ÍH as a whole). Analysis (C36H33N; O »0 .9 H20) C, H,?. (b) Example 20-4. { 3- [5- (4-Meti1-p-peraci-1-yl) -lfi-benzoim? Dazol-2-? L] -lfl-indazol-5-l] -isoquinoline: Anisole (229.4 mg, 2.12) was added mmol) to a solution of substance 20a (123.0 mg, 0.212 mmol) in glacial acetic acid (2.12 ral). Concentrated aqueous brohydric acid (2.12 ml) was added, and the mixture was heated to *, refluxing for 21 hours. After cooling, the reaction solution was added dropwise to a mixture that was a »*" 142 4"was rapidly stirring dichloromethane (50 ml), tetrahydrofuran (20 ml) and saturated aqueous sodium bicarbonate (30 ml), the layers were separated, and the organic layer X? washed with saturated aqueous sodium bicarbonate (20 ml), followed by water (20 ml) The organic layer was dried over magnesium sulfate, filtered, concentrated and purified by chromatography on silica gel (1: 20: 100). Concentrated aqueous NH 4 OH: EtOH: CH 2 C 12), which gave the slightly impure substance (76.0 mg, 78%) in the form of a red foam, The further purification by precipitation of dichloromethane / hexanes gave substance 20 in its pure state (47.1 mg, 48%) as a solid and pink color: Rf = 0.20 (1:20:50 concentrated aqueous NH4OH: EtOH: CH2C12); XH NMR (DMSO-de) [Some peaks double due to tautomeric isomerization] d 2.23 (á, 3H), 2.49 (m, 4H), 3.11 (m, 4H), 6.91 (m, 2H), 7.35 and 7.47 (2 d, ÍH as a whole, J = 9.0, 8.9 Hz), 7.61 (d, ÍH. J = 8: 9 Hz), 7.80 (m, 4H), 8.26 (d, ÍH, J - 7.7 Hz), 8.54 (s, 1H), 8.59 and 8.62 (2 s, HI as a whole), 9.39 (s, ÍH), 12.74 and 12.79 (2s, 1H as a whole), 13.73 and 13.76 (2s, ÍH as a whole). Analysis (C28H25N »0.7 H20) C, H, N. fix it 21: 2- [5- (3-H droxy-2-methyl-f-enyl) -lH-indazol-3-yl-l-yl-benzoijnidazol-4-ol - • (a) Intermediate 21a-2- [5-Iodo-l- (2-tpmethylsilanyl-ethoxyethyl) -lfl-indazol-3? l] -1-fibenzo-m-dazol-4-ol: The aldehyde 7b '(2.66 g, 6.62 mmol), and 2,3-diamylphenol (sold by Aldrich Chemicals) (822 mg, 6.62 mmol) were condensed in the presence of elemental sulfur in a manner analogous to the synthesis of intermediate 7c', what gave substance 21a (2.04 g, 61%) as a yellow solid: Rj = 0.15 (25% ethyl acetate in hexanes); H NMR (DMSO-de) [Some peaks double due to tautomeric isomerization] d-0.13 (s, 9H), 0.82 (t, 2H, J = 8.1 Hz), 3.59 (t, 2H, J = 7.8 Hz) , 5.85 (s, 2H), 6.59 (d, ÍH, J = 7.5 Hz), 7.01 (m, 2H), 7.71 (d, ÍH, J = 8.7 Hz), 7.81 (dd, ÍH, J = 8.8 , 1.5 Hz), 8.90 and 9.04 (2 s, ÍH as a whole), 9.49 and 9.74 (2 s, ÍH as a whole), 12.69 and 12.96 (2 s, ÍH as a whole). Analysis' (C20H23IN402S?) C, H, N. • * (b) Intermediate 21b-2- [5- (3-Methoxy-2? '• i * methyl-phenyl) -1- (2-trimethylsilanyl-ethoxymethyl) lH-indazol-3-yl] -lH-benzoimidazole- 4-ol: Boronic ester 9c '(250 mg, 1.01 mmol) and iodide 21a (510.6 mg 1.01 mmol) were coupled by the analogous procedure to the synthesis of intermediate 9d', which gave substance 21b (256.7 mg, 51%) in the form of a yellow foam Rf = 0¿2? (30% ethyl acetate in hexanes, mixed spots with substance 21a). 1H NMR (DMSO-de) [Some peaks double due to tautomeric isomerization] d -0.11 (s.9H), 0.85 (t, 2H, J = 8.1 Hz), 2.06 (s, 3H), 3.64 (t 2H , J = 7.7 Hz), 3. 85 (s, 3H), 5.90 (s, 2H), 6.55 (dd, ÍH, J = 7.2, 1.1 Hz), '6.96 (m, 4H), 7.26 (t, ÍH, J = 7.9 Hz), 7.46 ( dd, 1 H, J - 8.7, 1.5 Hz), 7.87 (d, 1H, J = 8.7 Hz), 8.40 and 8.55 (2 s, HI as a whole), 9.45 and 9.61 (2 s, HI as a whole), 12.62 and 12.91 (2 s, HI as a whole). Analysis (C28H32N4O3Si »0.4H20) C, H, N. (c) Intermediate 21c -2- [5- (3-Methoxy-2-methyl-phenyl) -lfi-indazol-3-yl] -lfi-benzoimidazole- 4-ol: Analogously to Example 3, treatment of substance 21b (174.5 mg, 0.349 mmol) with tetrabutylammonium fluoride gave 21c (59.8 mgr 46%) in the form of a whitish solid: Rf = 0.26 ( 5% methanol in dichloromethane); 1H NMR (DMSO-de) [Some peaks double ff * > • #; * • * 145 due to tautomeric isomerization] d 2.07 (s, 3H), B. 1 ^ (s, 3H), 6.53 and 6.62 (2 d, 1H as a whole, J = 7.4, 7.7'Hz), 6.96 (m, 4H), 7.26 (t, ÍH, J = 7.9 Hz), 7.37 (d , ÍH, J = B * S 1 * 'Hz), 7.66 (d, ÍH, J * 8.5 Hz), 8.35 and 8.49 (2 s, ÍH as a whole), 9.45 and 9.55 (2 s, ÍH as a whole), 12.53 and 12.78 (2 s, ÍH as a whole), 13.57 and 13.62 (2 s, 1H as a whole). '-! » HRMS calculated for C22H? 9N402 371.1508 (MH +), found 371.1523. '' * (d) EXAMPLE 21-2- [5- (3-Hydraxy-2-methyl-phenyl) -l-yldazol-3-yl] -lfi-benzoimidazole-4-ol: A Via a procedure analogous to synthesis of phenol 8 ', treatment of substance 21 c (45.9 mg, 0.124 mmol) with pipdin hydrochloride gave substance 21 (29.0 mg, 66%) in the form of a colored powder * toasted: Rf = 0.28 (10% methanol in dichloromethane); lE NMR (DMSO-de) [Some peaks double due to the,. tautomeric isomeptation] d 2.04 (s, 3H), 6.54 and 6.62 (dd and d, ÍH as a whole, J = 7.2, 1.3 and 7.7 Hz), 6.75 (d, 1H, J = 7.4 Hz), 6.85 (d, ÍH, J = 7.9 Hz) 7.01 (m, 3H), 7.37 and 7.38 '(dd and dd, 1H as a whole, J = 8.5, 1.5 Hz for each nail), 7.65 and 7.66 (2 d, 1H as a whole, J - 8.7 Hz for each), 8.35 and 8.48 (2 s, HI as a whole), 9.38 and 9.39 (2 s, 1H as a whole), 9.46 and 9.56 (2 s, HI as a whole), 12.52 and 12.77 * (2 s, ÍH as a whole), 13.55 and 13.60 (2 s, 1H as a whole) -.
HRMS calculated for C2, H.7N402 357.1351 (MH +), found 357.1360. Analysis (C2lH? 6N4O2 «0.8 CH3OH) C, H, N.
Ejeraplo 22: 6- (3-H? Drox? -prop? L) -2-methyl-3- [3- ((E - (a) Intermediate 22a-3-Am? no-2-met? l- -phenol: A suspension of 2-met? l-3-n? tro-phenol (Aldrich Chemicals) (29.8 g, 194.6 mmol) and Pallad or 10% carbon (3.01 g) in ethanol (350 ml) was stirred under hydrogen at a pressure of 40 psi for 3.5 hours. After filtration through a pad of celite, the solution was concentrated and purified by chromatography on silica gel (50% ethyl acetate in hexanes) to give 1a. aniline 22a (20.32 g, 85%) in the form of a colorless solid: Rf = 0.50 (50% ethyl acetate / hexanes); 1 H NMR (DMSO-de) d 1.87 (s, 3 H), 4.63 (s, 2 H), 6.08 (dd, 2 H, J-7.9, 10.5 Hz), 6.64 (t, ÍH, J = 7.9 Hz), 8.76 ( s, 1H). Analysis (C7H9NO) C, H, N. (b) Intermediate 22b-3-Yodo-2-methyl-phenol: 3-Amino-2-methyl-phenol 22a (18.35 g, 149 mmol) was diazotized in accordance with DeGraw method, et al. [DeGraw, J. I .; Brown, V. H .; Colwell, W. T .; Morrison, N.E., J. Med. Chem. , 17, 762 (1974)]. After performing column chromatography (10-50% ethyl acetate in hexanes), the aryl iodide 22b (9.06 g, 26%) was isolated in the form of an orange solid. Further purification by recrystallization of hexanes gave 5.63 g of pale orange needles: Rf = 0.35 (20% ethyl acetate / hexanes); E NMR (DMSO-de) d 2.22 (s, 3H), 6.80 (m, 2H), 7.24 (dd, ÍH, J = 7.5, 1.5 Hz), 9.75 (s, ÍH). (c) Intermediate 22c-1-Allyloxy-3-iode-2-methyl-benzene: Allyl bromide (1.57 g, 13.0 mmol) was added to a solution of 3-iodo-2-methylene phenol (2.026). g, 8.66 mmol) in acetone (18 ml). The solution was heated to reflux temperature for 2 hours, then cooled to room temperature, diluted with ethyl acetate (50 g.
The complex of bo-ranor ... dimethylsulfide (0.159 ml, 1.68 mmol borane) was added dropwise to a frozen solution (0 ° C) of intermediate 22d (459.8 nitrate *, ^, V.} 1.677 mmol) in dry ether (5.0 ml). The cooling P bath was removed, and stirring was continued for 1 hour. Absolute ethanol (2.5 ml) was added, followed by aqueous sodium hydroxide (2.5 N, 3.35 ml). The mixture was again cooled to 0 ° C, and hydrogen peroxide (30 wt% in H20, 0.27 ml) was added. After stirring at a temperature of 0 ° C for 15 minutes, the cooling bath was removed and the mixture was allowed to warm to room temperature for 1 hour. The solution was partitioned between ether (50 ml) and IN aqueous hydrochloric acid (final aqueous pH 2-3). The organic layer was dried over magnesium sulfate, filtered and concentrated in an orange oil. Purification by chromatography on silica gel provided alcohol 22e (353.1 mg, 72%) as a yellow oil Rf = 0.11 (20% ethyl acetate / hexanes); 1H NMR (DMSO-de) d 1.64 (quint, 2H, J = 7.0 Hz), 2.29 (s, 3H), 2.54 (t, 2H, J = 7.5 Hz) 3.39 (t, 2H, J = '6.5 Hz), 4.54, (br & * V 's, ÍH), 6.68 (d, ÍH, J - 8.1 Hz), 7.23 (d, ÍH, J = 8.1 Mz), v * 8.58 (s, 1H). (f) Intermediate 22f-6- (3-Hydroxypropyl) -2-methyl-3- [3- ((E) -styryl) -1- (2-trimethylsilanyl-ethoxymethyl) -lfi-indazole -5- il] -phenol: Aqueous sad carbonate solution (2M, 1.79 ml) was added to a degassed ester solution or j .QQ-i 16a (534.1 mg, 1.12 mmol), aryl iodide 22e (209.1 | 8J 0.716 mmol), and 1, 1-bis (diphenyl-1'-ferrocenedichloroprone) (II) (29 mg, 0.036 mmol) in DMF f >3.2 * ml) The mixture was heated in an oil bath at W? ß temperature of 80 ° C for 1.5 hours, then cooled and S ** - partitioned between ethyl acetate (50 ml) and water (10 ml). ml) The organic layer was dried over magnesium sulfate, filtered and concentrated, purification by chromatography on silica gel (20 to 50% ethyl acetate in hexanes) gave I the substance 22f (301.9 mg, 82%). ) as a yellow foam: Rf = 0.07 (20% ethyl acetate / hexanes); XH MBfc (DMSO-de) 6.-0.09 (s, 9H), 0.83 (t, 2H, J = 7.9 Hz), 1.73 (qumt, 2H, J = 7.5 Hz), 2.09 (s, 3H), 2.65 (t, 2H, J - 7.5'-Hz), 3.46 (t, 2H, J - 6.5 Hz), 3.58 (t , 2H, J = 8.0 H¿), 5.78 (s, 2H), 6.74 (d, ÍH, J = 7.7 Hz), 6.98 (d, ÍH, J = 7.9 Hz), 7.27 (m, ÍH), 7.37 ( ra, 3H), 7.56 (m, 2H), 7.73 (m, 3H), 8.06 (s, 1H), 8.25 (br s, 1H).
CH2C12) C, H, N. (g) Intermediate 22 - 6- (3-Hi | dr3? I-propyl) -2-methyl-3- [3- ((E) -styryl) -1 phydazole -5-ilJ-feaolr Substance 22 was prepared in a similar way to • Example 3, the treatment of the intermediate substance? £ * * (202.9 mg, 0. 394 mraol) with tetrabutylamine fluoride?? Ig g? •H.H. f < rr Substance 9a ', intermediate 23a (3.16 g, 18.8 mmol) was hydrogenated in ethanol (300 ml) to. provide 23 (2.23g, 86%) in the form of a yellow-brown solid. The subsequent purification by recrystallization * P--. of ethanol resulted in substance 23b (1.04 g ,: 40%). • X in the form of yellow needles: Rf = 0.17 (75% acetate "' -2 ethyl in hexanes); ? E NMR (DMSOd6) d 4.36 (br s, -6H), 4.90 (br s, ÍH), 6.42 (m, 3H). Analysis (CH? 0N2O) C, H, N. "(c) Intermediate substance 23c. { 2- [5-iodo-l * | 2-trimethylsilanyl-ethoxymethyl) -lfí- '' inñ3t¿ol-3-xl í-% IÍX? benzoimidazol-4-yl} -methanol: ? "Í # M:;« «- A- Substance 23c was prepared in a manner similar to the synthesis of substance 7c '. Condensation of the diamine to "23b (587.3 mg, 4.25 mmol) with the aldehyde 7b '(1.71 g, 4.25 tf'r * ramol) in the presence of elemental sulfur gave the substance 23c (1.57 g, 71%) in the form of a solid of color A yellow:? E NMR (DMSO-de) [Some peaks double due to tautomeric isomerization] d.-0.13 (s, 9H), 0.82 (t, yA ... 2H, J * 7.7 Hz ), 3.58 (t, 2H, J = 7.9 Hz), 4.87 (br s, ÍH), 5.04 (br s, ÍH), 5.22 (br s, 1H), 5.87 (s, 2H), 7.26 (, 2H) , 7.39 and 7.67 (m and br s, ÍH in conjunction), 7.75 (d, 1H, J = 8.7 Hz), 7.83 (dd, ÍH, J = 8.8, 1.5 Hz), 8.95 (d, ÍH, "J - 1.1 Hz), 12.97 and 13.13 (2 s, ÍH as a whole). Analysis (C2? H25IN02S?) C, H, I, N. (d) Intermediate 23d-2-Met? L-3- (4, 4, 5,5- tetramethyl- [1,3,2] dioxaborolan- 2-? L) -phenol: Through a synthetic method analogous to the synthesis of substance 9c, the iodide 22b (1.21 g, 5.17 mmol) was converted to the boronic ester 23d (1.15 g, 95%), a solid crystalline white: Rf = 0.18 (10% ethyl acetate in hexanes); XH NMR (CDC13) d 1.35 (s, 12H), 2. $, * * (s, 3H), 6.87 (dd, 1H, J «7.9, 1.0 Hz), 7.08 (t, ÍH, J - 7.5 Hz) , 7.35 (dd, ÍH, J = 7.4, 1.1 Hz). Analysis (C13H19BO3 »0.2 H20) C, H. (e) Intermediate 3e-3- [3- (4-Hydroxymethyl-1-yl-benzoimidazol-2-yl) -1- (2-tr? methylsilane-ethoxymethyl) -lfi-indazol-5-yl] -2- methyl-phenol: Substance 23e was prepared in a manner similar to the synthesis of substance 9d '. Iodide 23c (276.3 mg, 0.514 mmol) and boronic ester 23d (300 mg, 1.28 mmol) were coupled to give substance 23e (128.2 mg, 50%) as a yellow solid: Rf = 0.16 (40%) ethyl acetate in hexanes); XH NMR (DMSO-d6) [Some peaks double due to tautomeric isomerization] d.-O.ll »(s, A 9H), 0.85 (t, 2H, J = 7.9 Hz), 2.03 and 2.07 (2 s, 3H as a whole), 3.63 (t, 2H, J = 7.7 Hz), 4.87 and 4.97 (2 d, 2H as a whole) , J = 5.8 and 5.5 Hz), 5.11 and 5.25 (2 t, ÍH as a whole, J - 5.6 and 6.1 Hz), 5.92 and 5.93 (2 s, 2H as a whole), 6.76 (dd, ÍH, J = 7.5, 3.4 Hz), 6.86 (d, 1H, J = 7.9 Hz), 7.17 (m, 3H), 7.39 and 7.60 (dd and d, ÍH as a whole, J = 6.8, 2.1 and 7.9 Hz), 7.49 (d, ÍH, J - 8.7 Hz), 7.89 (d, ÍH, J = 8.9 Hz), 8.44 and 8.47 (2 s, 1H as a whole), 9.46 and 9.48 (2 s, ÍH as a whole), 12.91 and 13.09 (2 s, ÍH on the whole) . Analysis (C28H32N03S? »0 .3H20) C, H, N. (f) Example 23 -3- [3- (4-Hydroxymethyl-1-yl-benzoimidazol-2-yl) -lfi-indazol-5-yl ] -2-methyl-phenol: The substance of Example 23 was prepared in a manner similar to Example 3. The treatment of substance 23e (130.7 mg, 0.261 mraol) with tetrabutylammonium fluoride é * »> * 155 gave substance 23 (61.6 mg, 64%) as a white solid: Ry = 0.22 (70% ethyl acetate and hexanes); XH NMR (DMS0-d6) [Some peaks double due to / * to the tautomeric isomerization] d 2.04 and 2.07 (2 s, 3H tn set), 4.86 and 4.97 (2 d, 2H together, J = 6.0 and 5.7 2 Hz), 5.10 and 5.23 (2 t, ÍH as a whole, J = 5.6 and 6.0 Hz), 6.76 (d, ÍH, J = 7.2 Hz), 6.85 (d, ÍH, J «8.1 Hz), 7.14 (m, 3H), 7.37 and 7.58 (dd and d, ÍH as a whole, J = 7.2, 1.9 and 7.7 Hz), 7.40 (dd, ÍH, J = 8.5, 1.5 Hz), 7.67 (d, ÍH, J - 8.1 Hz ), 8.39 and 8.42 (2 s, HI as a whole), 9.43 and 9.45 (2 S, 1H as a whole), 12.81 and 12.96 (2 s, HI as a whole), 13.65 and 13.70 (2 s, HI as a whole). Analysis (C224H? 8N402 * 1 .0 CH3OH) C, H, N.
Example 24: 7- [3- ((E) -Estyril) -lfí-lnda ^ ol-5- l] -isoquinolipa * < * '> ^ [Pyridme = Pipdma, Dioxane = Dioxane] J = 8.4, 1.8 Hz), 8.51 (m, 3H), 9.41 (s, 1H), 13.28 (s, ÍH). Analysis (C24H1 N3 «0.6 CH2OH) C, H, N.
Example 25: 4- [3- (1-Benzoimidazol-2-yl) -lfl »indaz; Dl-5-yl] - ', isoquinoline (a) Intermediate 25a-3- (1-Benzoimidazol-2-yl) -5- (4, 4, 5, 5-tetramethyl- [1,3,2] -dioxaborolan-2-yl) -1- ( 2-trimethylsilanyl-ethoxymethyl) -lfi-indazole: Through a procedure analogous to the synthesis of the boronic ester 19e, the iodide 7c '(2.36 g, 4.81 mmol) was converted to boronic ester 25a (1.43g, 61% ), a white crystalline solid: lE NMR (DMSO-de) 5.-0.13 (s, 9H), 0.82 (t, 2H, J = 7.7 Hz), 1.35 (s, 12H), 3.59 (t, 2H , J --7.9 Hz), 5.89 (s, 2H), 7.24 (m, 2H), 7.53 (m, ÍH), 7.83 (m,, 3H), 8.95 (s, ÍH), 13.15 (s, ÍH). Analysis (C2eH35B 4? 3Si) C ^ H, N. * (b) Intermediate 25b-4-t3- (10- ^ Ben2? Imidazol-2-yl) -1- (2-tpmethylsilanyl-ethoxymethyl) -1H- mdazol-5-? l] -isoquinoline: Through a procedure analogous to the synthesis of substance 19f, 4-bromoisoquinolma (238 rag, 1.14 mmol) was coupled with boronic ester 25a (280.4 mg, 0.572 mmol) to provide 25b (237.5 mg, 84%) as a white solid: Rf = 0.20 (50% ethyl acetate in hexanes); 1 H NMR (DMSO-de) 5.-0.07 (s, 9H), 0.86 (m, 2H), 3.67 (t, 2H, J = 7.9 Hz), 5.98 (s, 2H), 7.20 (br m, 2H) , 7.55 (br m, ÍH), 7.65 (br m, ÍH), 7.71 (m, 4H), 8.07 (d, ÍH, J = 8.7 Hz), 8.27 (dd, ÍH, J- 7.2, 1.7 Hz), 8.56 (s, ÍH), 8.66 (d, ÍH, J = 0.8 Hz), 9.41 (s, ÍH), 13.17 (s, ÍH). (s) Example 25-4- [3- (1-Benzoimidazol-2-yl) * 1-X-ndazol-5-yl] -isoquinoline: Substance 25 was prepared in a manner similar to that of Example 3. Intermediate 25b (152.4 mg, 0.310 mmol) was treated with tetrabutylammonium fluoride to give substance 25 (61.9 mg, 55%) as a white foam: Rf = 0.16 (70% ethyl acetate in hexanes); XH NMR (DMSO-de) d 7.18 (br m, 2H), 7.56 (br m, 2H), 7.63 (dd, ÍH, J = 8.5, 1.7 Hz), 7.81 (m, 4H), 8.27 (dd / ÍH, J 1H), 1 Example 26: 3- [3- (l-Benzo-m-dazol-2-ll) -lfi-mdazoI- (a) Intermediate 26a-3- [3- (l-Benzo-mrdazole-2-? l) -1- (2-tmetmet? ls? lan? l-ethox? met? l) -lfl-ndazole- 5-?] -6- (3-hydroxy? -prop?) -2-methyl-1-phenol: Via a procedure analogous to the synthesis of substance 25b, boromco-25a ester (303 mg, 0.618 mmol) with iodide 22e (180.5 mg, 0.618 mmol), which gave substance 26a (124.4 mg, 38%) as a white solid: R; = 0.30 (50% ethyl acetate in hexanes); * H NMR (DMS0-d6) d -0.11 (s, 9H), 0.85 (t, 2H, J «7.9 Hz), 1.74 (quint, 2H, J = 7.0 Hz), 2.08 (s, 3H), 2.66 ( t, 2H, J = 7.7 Hz), 3.47 (q, 2H, J - 5.3 Hz), 3.63 (t, 2H, J = 7.9 Hz), 4.60 (t, ÍH, J »5.0 Hz), 5.91 (s, 2H), 6.75- ** 4k (d, ÍH, J = 7.7 Hz), 7.01 (d, ÍH, J = 7.7 Hz), 7.20. { quint ', 2H, J = 8.1 Hz), 7.49 (m, 2H), 7.71 (d, 1H, J = 7.7 Hfc), / 7.88 (d, 1H, J = 8.7 Hz), 8.33 (s, ÍH), 8.42 (s, ÍH), 13. H (s, ÍH). Analysis (C3oH36N4? 3S? "0.6 ethyl acetate) C, H, N. * * (b) Example 26 -3- [3- (1-Benzoimide = ol-2-yl) -liimidazole-5-yl ] -6- (3-hydroxy-propyl) -2-methyl-phenol: Through a procedure analogous to Example 3, deprotection of 26a (99.4 mg, 0.188 mmol) with tetrabutylammonium fluoride provided substance 26 (26.9 m < |, 36%) in the form of a white solid: = 0.19 (70% ethyl acetate in hexanes); 1 H NMR (DMSO-d 6) 6 1.74 (quint, 2 H, J - 7.4 Hz), 2.08 (s, 3 H), 2.66 (t, 2 H, J = 7.4 Hz) * 3.47 (q, 2 H, J - 5.1 Hz) , 4.59 (t, ÍH, J - 5.1 Hz), 6.74 (d, 1H, J - 7.7 Hz), 7.00 (d, ÍH, J = 7.7 Hz), 7.19 (quint, 2H, J = 7.9 Hz), 7.38 (dd, 1H J = 8.5, 1.5 Hz), 7.50 (d, 1H, J = 7.4 Hz), 7.67 (m, 2H), 8.30 (s, ÍH), 8.37 (s, ÍH), 12.96 (S, ÍH) ), 13.66 (s, ÍH). Analysis (C 24 H 22 N 2"0.4 ethyl acetate) C, H, N.
Example 27: 1- [3- ((E) -Estipl) -lfl-indazol-5-yl] -piperidii | H4- ol [Imidazole = Imidazole] (a) Intermediate 27a-4- (tert-Butyl-dimethyl-silanyloxy) -piperidine: Imidazole (4.18 g, 61.4 mmol), 4-hydroxypiperidine (2.07 g, 20.46 mmol) were dissolved, and chloride of tert-but-dimethylsilyl (4.63 g, 30.7 mmol) in diechloromethane (50 ml) and stirred at a temperature of 23 ° C for 4 hours. The mixture was then washed with saturated aqueous sodium bicarbonate solution (3 x 50 ml) and water (50 ml), dried over magnesium sulfate, filtered and concentrated under high vacuum to provide 27a (2.60 g, 59% ) as a yellow oil that crystallizes when at rest: XH NMR (CDCI3) d 0.05 (s, 6H), 0.90 (s, 9H), 1.46 (m, 2H), 1.81 (m, 2H), 2.71 (m, 2H), 3.09 (m, 3H), 3.77 (septet, ÍH, J = 3.9 Hz). Analysis (CnH25NOS? * 0.2 C H2C12) C, H, N. (b) Intermediate substance 27b - §-. { 4-. { (Dimefcyi-ethyl) -dimethyl-silanyloxy] -piperidin-1-yl} -3- ((E) -styryl) -1- (2-trimethylsilanyl-ethoxymethyl) -Ify-indazole: Sodium tert-butoxide (163 mg, 1.70 mmol), tris (dibenzyl-idinacetone) dipalladium (0) were added. (26 mg, 0.03 * mmol), and CyMAP-1 (see Oíd et al., J. Am. Chem. Soc., 120, 9722 (1998) regarding the structure of this ligand) (33 mg, 0.085 ramol) to a degassed solution of substance 27a (241.1 mg, 1.12 mmol) and iodide 7a '(269.3 mg, 0.565 mmol), in ethylene glycol dimethyl ether (DME, 2.0 ml), The mixture was heated in an oil bath at a temperature of 80 ° C for 17 hours. After cooling to room temperature, the mixture was diluted with ethyl acetate (50 ml) and filtered to remove the black precipitate. The filtrate was washed with water (10 ml) and saturated aqueous sodium chloride (10 ml), dried over magnesium sulfate, filtered, concentrated, and purified by chromatography on silica gel (10 to 50% ethyl acetate). ethyl in hexanes) to provide the substance 27b (177.7 mg, 56%) as an orange oil: Rf = 0.28 (20% ethyl acetate in hexanes); XE NMR (CDC13) d. ~~. 06 (s, 9H), 0. 09 (s, 6H), 0.90 (m, 2H), 0.92 (s, 9H), 1.80 (m, 2H), 1.97 (m, 2H), 3.07 (m, 2H), 3.44 (m, 2H), 3.58 (t, 2H, J - 8.4) Hz), 3.92 (m, ÍH), 5.69 (s, 2H), 7.29 (m, 2H), 7.41 (ra, 6H), 7.61 (d, 2H, J = 8.7 Hz). 164 1 4 * (c) Example 27-1- [3- ((E) -Styryl) -l-mdazol-5-yl] -piper? din-4-ol: Through a procedure analogous to that of Example 3, the treatment of intermediate 27b (121.4 mg, 0.22 mmol) with tetrabutyllaubstituted fluoride gave 27 (33.1 mg, 47%) as a yellow foam: Rf = 0.15 (70% ethyl acetate / hexanes); 1 H NMR (DMSO-de) d 1.55 (m, 2H), 1.86 (m, 2H), 2.83 (m, 2H), 3.47 (m, 2H), 3.61 (m, 1H), 4.68 (d, ÍH, J = 4.2 Hz), 7.22 (m, 2H), 7.37 (m, 5H), 7.55 (d, ÍH, J = 16.5 Hz), 7.69 (d, 2H, J - 7.2 Hz), 12.89 (s, ÍH). Analysis (C2CH21N3O «0.4 H20« 0.4 ethyl acetate) C, H, N.
Example 28: l- [3- ((E) -Est r l) -lfí ~ ndazol-5- »i3.3 *. pipe? d? n-3-ol [Imidazole = Imidazole] (a) Intermediate 28a-3- (tert-Butyl-dimethyl-silanyloxy) -piperidine: - (181.5 mg, 0.322 mmol) with tetrabutylammonium fluoride gave substance 28 (47.6 mg, 46%) as a yellow foam: R, = 0.19 (70% ethyl acetate / hexanes); * H NMR (DMSO-de) d 1.34 (m, 1H), 1.70 (m, 1H), 1.95 (m, 2H), 2.55 (m, 1H), 2.72 (mIH), 3.46 (m, ÍH), 3.63 (m, ÍH), 3.74 (m, 1H), 4.88 (d, ÍH, J - 4.5 Hz), 7.24 (dd, ÍH, J = 9.0, 1.8 Hz), 7.33 (t, ÍH, J = 7.2 Hz ), 7.42 (m, 5H), 7.63 (d, ÍH, J = 16.5 Hz), 7.76 (d, 2H, J = 7.2 Hz), 12.97 (s, ÍH). Analysis (C20H2? N30 »0.3 H20) C, H, N.
Example 2 »: [2- (5-Isoquinol? N-4-? L-l-nidazol-3? L) -lfi-benzoim dazol-4-yl] -methanol (a) Intermediate substance 29a -. { 2- [5- (4, 4, 5, 5-Tetramet? L- [1, 3, 2] d? Oxaborolan-2? L) -l- (2- -. trimethylsilanyl-ethoxymethyl) -lfl-indazol-3-yl] -1K-benzoimidazol-4-yl} -methanol: Through the use of a procedure similar to the synthesis of I9e boronic ester, iodide 23c (512.8 mg, -0,985 mmol) was converted to boronic ester 29a (312.0 mg, 61%), a white foam: Rf = 0.28 (5% methanolic in dichloromethane); XH NMR (DMSO-d6) [Some peaks double due to tautomeric isomerization] d.-0.13 (s, 9 H), 0.83 (t, 2H, J = 7.7 Hz), 1.35 (s, 12H), 3.60 ( t, 2H, J = 8.1 Hz), 4.87 (br s, ÍH), 5.06 (br s, ÍH), 5.24 (m, ÍH), 5.90 (s, 2H), 7.26 (m, 2H), 7.40 and 7.71 (2 d, ÍH as a whole, J = 7.2 and 7.9 Hz), 7.82 (m, 2H), 8.95 (s, 1H), 12.93 and 13.10 (2 s, ÍH as a whole). Analysis,. (C27H37BN4O4Si »0.5 H20) C, H, N. b) Intermediate substance 29b-. { 2- [5-isoquinolin-4-yl-1- (2-trimethylsilanyl-ethoxy-methyl) -lfl-indazol-3-iD-lyn-benzoimictezol-4-yl} -methanol: Through the use of a procedure similar to the synthesis of the substance 19f, 4-bromoisokoline (193 mg, 0.927 mmol) was coupled with boronic ester 29a (241.2 mg, 0.463 mmol) to provide 29b (171.1 mg, 71%). %) in the form of a white foam: R = 0.22 (75% ethyl acetate in hexanes); XH NMR (DMSO-de) [Some peaks double due to tautomeric isomerization] 5.-0.08 (s, \ 9H), 0.88 (t, 2H, J = 7.7 Hz), 3.68 (t, 2H, - J = 7.9 H ^), r. .. *. * & ,, "168 4. 88 (d, 2H, J = 5.3 Hz), 5.05 and 5.25 (2 br, ÍH in P set), 5.98 (s, 2H), 7.22 (m, 2H), 7.40 and 7.57 (2m, ÍH "together ), 7.77 (m, 4H), 8.07 (d, 1H, J = 8.5 Hz), 8.27 ^ (dd, 1H, J = 7.2, 1.5 Hz), 8.57 (s, ÍH), 8.70 (br s, 1H) , • **. 9.41 (s, ÍH), 12.97 and 13.14 (2 s, ÍH as a whole) Analysis (C3? H31N5? 2Si «0.4 H20) C, H, N. (c) Example 29- [2- (5-Isoquinolin-4-yl-lfl-inda2dl-s »«, 3-yl) -lfi-benzoimidazol-4-i1] -methanol: Substance 29 was prepared in a manner similar to that of Example 3. The treatment of Intermediate 29b (129.0 rag, 0.247 mmol) with tetrabutylammonium fluoride provided 29 (58.3 mg, 60%) in the form of white powder: XH NMR (DMSO-de) [Some peaks double because of, the tautomeric isomerization] d 4.88 (t, 2H, J = 6.2 Hz), 5.03 and 5.23 (2 t, ÍH as a whole, J = 5.6 and 6.2 Hz), 7.20 (m, 2H), 7.38 and 7.53 (myd, ÍH in set, J = 7.4 Hz ta the doublet), 7.63 (dd, ÍH, J - 8.7, 1.3 Hz), 7.82 (m, 4H, 8.27 (d, ÍH, J = 7.4 Hz), 8.55 (s, ÍH), 8.63 and 8.66 (2 s, 1H as a whole), 9.40 (s, ÍH), 12.87 and 13.02 (2 s, ÍH as a whole), 13.81 and 13.86 ( 2 s, HI as a whole). Analysis- (C24H17N5O »0 .4 H2O« 0 .3 CH2C12) C, H, N. %.
Example 30: 2 - [2 - (5-Isoquinolin-4-yl-l-yn-2-yl-3-yl) -, - 1-yl-benzoimidazol-4-yl] -ethanol? i _ . * * t * BS & A "X- 169 (a) Intermediate 30a-2-. { 2- [5- * Iso-ainolin-4-yl-1- (4-methoxy-benzyl) -lfi-indazol-3-yl} -lfi-benzoimidazol-4-yl} Ethanol: A suspension of 10% carbon palladium (66 mg) and 2- (2-amino-3-nitrophenyl) ethanol [see Seno, K ^ oru; Hagishita, Sanji; Sato, Tomohiro; Kuriyama, Kaoru; J. Chem. Soc. Perkin Trans. 1; 2012 (1984) regarding the synthesis of, * - this reagent] (531.5 mg, 2.92 mmol) at all etanc * l (50 ml) was stirred under nitrogen at a pressure of 40 psi for t 3 hours. After filtration and concentration, crude 2- (2,3-diaminophenyl) ethanol (474.4 mg) was obtained in the form of a red oil, which crystallized on standing: f = 0.08 (75% ethyl acetate in hexanes); XH NMR (DMSO-de) 5 2.58 (t, 2H, J = 6.9 Hz), 3.53 (t, 2H, J * 7.2 Hz), 4.32 (br s, 5 H), 6.29 (m, 2H), 6.40 ( dd, ÍH, J = 6.9, 2.1 Hz). Without further purification, this crude diamma was condensed to aldehyde 19f (1.10 g, 2.81 mmol) in the presence of sulfur, similar to the synthesis of intermediate 7c ', to provide substance 30a (930.8 mg, 63%) as a yellow foam: R = 0.19 (ethyl acetate); XH NMR (DMSO-de) [Some peaks double because of the Example 31: [2- (5-Isoquinol-4-yl-l-yl-mctazol-3-yl) -lfl-benzoimidazol-4-ylmethyl] -dimet-l -amine? [Anisol = Anisole] (a) Intermediate 31a - (2- [5-Isoquinolin-4-? Ll- (4-methox? -benzyl) -lfl-ndazol-3? L] -lfi-benzoimidazole - '' r 4-íl.}. Methanol: Through a synthesis similar to the synthesis of substance 19h, the aldehyde 19f (3.67 g, 9.33 mmol) and diamma 23b (1.29 g, 9.33 mmol) were condensed in the presence of sulfur to provide 31a (2.60 g, 54%) as a yellow solid: Rf = 0.19 (^ 5% ethyl acetate in hexanes); XH NMR (DMS0-d6) [Some peaks double because to tautomeric isomerization] 5 3.71 (s, 3H), 4.88 (d, 2H, J = 5.5 Hz), 5.04 and 5.25 (2 t, 1H as a whole, J = 5.6 and 6.1 Hz), 5.81 and 5.83 (2 s) , 2H in ^ - set), 6.93 (d, 2H, J = 8.5 Hz), 7.21 (, 2H), 7.38 and 7.54 (2 d, 3H together, J = 7.4 and 7.5 Hz), 7.66 (d, ÍH, J = 8.7 Hz ), 7.77 (m, 3H), 8.01 (dd, ÍH, J = 8.7, 4.0 Hz), 8.26 (d, ÍH, J = 7.7 Hz), 8.54 and 8.55 (2 s, HI as a whole), *** »8.65 and 8.68 (2 s, ÍH as a whole), 9.39 (s, ÍH), 12.88 and 13.05 (2 s, ÍH as a whole). Analysis (C32H25N5? 2 * 0.3H20) C, H, N. (b) Intermediate 31 b- (2- [5-isoquinolin-4-yl-l- (-methoxy-benzyl) -lfi-indazol-3 -yl] -lfl-benzoimidazol-4-methylmethyl] -dimethyl-amine: Sulphonyl methane chloride (119.3 mg, 1.04 mmol) was added dropwise to a solution of 31a (527.5 mg., 1.03 mmol) and diisopropylethyl amine (153.3 mg, 1.19 mmol) in ^ tetrahydrofuran (12.0 ml), was cooled to a temperature of 0 ° C in an ice bath. After stirring at 0 ° C for 2.5 hours, the beaker of the reaction was conditioned with a digital condenser cooled with dry ice, and the dimethyl amine gas was condensed in the reaction solution until the volume grew in an level of about 5 ml. The stirring was continued at a temperature of 0 ° C for 4 hours, then at room temperature for 15 hours. The mixture was partitioned between ethyl acetate (100 ml) and saturated aqueous sodium bicarbonate solution (20 ml). The organic extracts were dried over magnesium sulfate, filtered, s * e V 173 concentrated, and columned (silica gel, 5 to 10% ethanol 'in dichloromethane), to provide 31b (250.2 mg, 45%) in the form of a pale yellow solid: Rf (10% methanol in dichloromethane); XH NMR (DMSO-de) d 2. 5 6H), 3.71 (s, 3H), 3.84 (s, 2H), 5.83 (s, 2H), 6.93 (d, 21,, J = 8.7 Hz), 7.14 (m, 2H), 7.36 (d, 2H) , J - 8.5 Hz), 7-48 (m, 1H), 7.67 (d, ÍH, J - 8.9 Hz), 7.77 (quintet, 2H, J - 6.4 Hz), 7.89 (m, ÍH), 8.00 (d , ÍH, J = 8.9 Hz), 8.26 (d, ÍH, J = 7.5 Hz), 8.55 (s, ÍH), 8.71 (s, ÍH), 9.39 (s, 1H) \ 10 13.03 (br s, ÍH) . Analysis (C34H3oNeO "1 .1 H20) C, H, N. (c) Example 31 - [2- (5-Isoquinolin-4-yl-l-yl-indazol-3-yl) -l-benzoimidazol-4-ylmethyl] - dimethylamine: A mixture of 31b (125.7 mg, 0.233 mmol), anisole (252 mg, 2.33 mmol), trifluoroacetic acid (2.3 ml), and concentrated sulfuric acid (0.2 ml) was stirred at room temperature for 66 hours , then added dropwise to a rapidly stirring mixture of saturated aqueous sodium bicarbonate (75 ml) and ethyl acetate (25 ml). The layers were separated and the aqueous layer was extracted with ethyl acetate (2 x 50 ral). The combined organic layers were dried over magnesium sulfate, filtered, concentrated, and purified by chromatography on silica gel (25 to 40% methanol in dichloromethane) to give 31 (35.8 mg, 5 37%) in the form of powder of white color: Rf = 0.09 (10% JM-, * * - 174 methanol in diechloromethane); H NMR (DMSO-de) [Some peaks double due to tautomeric isomerization] d 2.15 and 2.21 (2 br s, 6H as a whole), 3.80 (s, 2H), 7.12 (br s, 2H), 7.40 and 7.54 (2 m, ÍH as a whole), 7.64 (d, ÍH, J = 9.0 Hz), 7.83 (m, 4H), 8.26 (d, ÍH, J - 7.5 Hz), 8.55 (s, ÍH), 8.63 and 8.73 (2 brs, ÍH as a whole), 9.39 (s, 1H), 13.02 (br s, ÍH), 13.83 (br s, ÍH). Analysis (C26H22 6 «0.7 H2O» 1.0 CH3OH) C, H, N.
.Example 32: [2- (5-Isoquinol n-4- l-l-nidazol-3-i3,) -Líf- * benzom? Dazol-4-? Lmet l] -met l-amine [Anisol = Anisol] (a) Intermediate substance 32a-. { 2- [5-Isoquinoline-4- * 1-l- (4-methox? -benzyl) -lfl-ndazol-3-? L] -lfi-benzo? M? Dazo |.-4-limethyl} -methyl-amine: X- Through the use of a procedure similar to the synthesis of substance 31b, alcohol 31a (516.6 μg, 1.01 mmol) was treated with methanesulfonyl chloride and dusopropylethyl amine at a temperature of 0 °. C for 1 hour. Instead of a condensed gas, a "*" * 175 * solution of methylamine in tetrahydrofuran (2.0 M, 5-.0) and stirring was continued at ambient temperature for 15 hours.The extractive treatment and the # silica gel chromatography similar to those applied to substance 31b gave analogue mono-methyl to (170.5 mg, 32%) in the form of a whitish solid: Rf = 0.16 (1: 20: 300 conc. Aqueous NH40H: ethanol: dichloromethane); X NMR (DMSO-de) d 2.26 (S, 3fi), 3.71 (s, 3H), 4.03 (s, 2H), 5.82 (s, 2H), 6.93 (d, 2H, J - 8.7 Hz), 7.14 (d, 2H, J = 4.7 Hz), 7.37 (d, 2H, J * ' 8. 7 Hz), 7.46 (m, ÍH), 7.67 (dd, ÍH, J - 8.7, 1.3 Hz), 7.77 (m, 2H), 7.89 (d, ÍH, J = 7.7 Hz), 8.01 (d, ÍH, J * 8.5 Hz>, 3.25 (dd, ÍH, J - 7.0, 1.8 Hz), 8.55 (s, ÍH ), 8.68 (s, 1H), " 9. 39 (s, 1H). Analysis (C33H28NeO »0.6 H20) C, H, N. (b) Example 32 - [2- (5-? Soqu? Nolin-4 ~ al-lIf ~ indazol-3? L) -lfí-benzo? M? dazol-4-? lmet? l] -myl-amine: Deprotection through the use of a procedure similar to the synthesis of 31 provided the substance 32 (47.5 mg, 63%) in the form of a foam d @ color whitish: Rf = 0.29 (1: 20: 100 NH40H concentrated water: ethanol: dichloromethane); XH NMR (DMSO-de) d 2.30 (s, 3H), 4.07 (s, 2H), 7.15 (d, 2H, J = 4.5 Hz), 7.47 (A, ÍH), 7.64 (dd, ÍH, J * 8.5 , 1.5 Hz), 7.83 (m, 4H), 8.26 (d, 'ÍH, J - 7.2 Hz), 8.56 (s, ÍH), 8.66 (s, ÍH), 9.39 (s, IB) • Analysis (C25H2oN6 «1.0EtOH« 0.2 hexanes) C, H, N. 1H), 9.39 (s, ÍH), 13.03 (br s, ÍH). Analysis (C36H32N6? »0.2 CH2C12) C, H, N. (b) Example 33-4- [3- (4-Pyrrolidin-1-ylmethyl * lJfc >; benzoimidazol-2-yl) -lfl-indazol-5-yl] -isoquinoline: A solution of 33a (109.2 mg, 0.193 mmol) in ^ 5% concentrated sulfuric acid / trifluoroacetic acid (2.0 ml) was stirred at room temperature for 20 minutes. hours, then added dropwise to a rapidly stirring mixture of tetrahydrofuran (25 ml), and saturated aqueous sodium carbonate (25 ml). Ethyl acetate (25 ml) and water (15 ml) were added, and the layers were separated. The aqueous layer was extracted with ethyl acetate (3 x 50 ml), and the combined organic fractions * were dried over magnesium sulfate, filtered and concentrated. Purification by chromatography on silica gel (1: 20: 100 conc. Aqueous NH 4 OH: ethanol dichloromethane) provided 33 (25.4 mg, 30%) in the white powder form: 1 H NMR (CD3OD) d 1.77 (br s) , 4H), 2.69 (br s, 4H), 4.12 (s, 2H), 7.24 (d, 2H, J «4.0 Hz), 7.80 (m, 5H), 8.06 (d, ÍH, J = 7.9 Hz), 8.23 (d, ÍH, J = 7.5 Hz), 8.53 (s, ÍH), 8.69 (s, 1H), 9.29 (s, ÍH). Analysis (C28H24Ne «0.9 MeOH) C, H, N.
Example 34: 4-. { 3- [4- (2-Pyrrolidin-1-yl-ethyl.) - lB- - 'benzoimida-2-yl-2-yl] -lfl-indazol-5-yl) -isoquinoline t m > & Four . 178 (a) Intermediate substance 34a-4-. { l- (4-MetFxi- * 'benzyl) -3- [4- (2-pyrrolidm-l-yl-ethyl) -l, l-benzoimidazole-2-? l] -lfl-ndazol-5-? } - Soquínolina: With a similar procedure to the synthesis of substance 33a, alcohol 30a (441.5 mg, 0.84 ramol) was • made into the substance 34a (204.6 mg, 42%), a whitish foam: Rf = 0.08 (1: 20: 400 concentrated aqueous NH 4 OH: ethanol: dichloromethane); XH NMR (DMSO-de) d 1.38. (br s, 4H), 2.31 (br s, 4H), 2.79 (m, 2H), 3.07 (m, 2H), 3.71 (s, 3H), 5.81 (s, 2H), 6.93 (d, 2H, J = 8.8 Hz), 6.98 (d, ÍH, J = 7.2 Hz), 7.08 (t, ÍH, J = 7.7 Hz), 7.36 (d, 2H, J = 8.7 Hz), 7.39 (m, 1H), 7.66 ( dd, ÍH, J = 8.5, 1.5 Hz), '7.76 (m, 2H), 7.89 (d, ÍH, J = 7.7 Hz), 8.01 (d, ÍH, J = 8.7 Hz), 8.25 (d, 1H, J = 7.4 Hz), 8.53 (s, 1H), 8.75 (br s, 1H), 9.38 (s, ÍH), 13.00 (br s, ÍH). Analysis (C37H34N6O »0.6 H20) C, H, N. (b) Example 34-4. { 3- [4- (2-Pyrrolidin-1-l-ethyl) -l, -benzo-m-dazol-2-yl] -lfl-ndazol-5-l} Isoquinoline: Substance 34 was prepared in a similar manner to example 33. The treatment of substance 34a (66.2 mg, 0. 114 mmol) with trifluoroacetic acid / sulfuric acid (© the ratio of 3: 1) gave substance 34 (24.7 mg ^ 47%) in the white powder form: Rf = 0.38 * (1: 20: 100 aqueous NHOH) concentrate: ethanol: dichloromethane); "H NMR (DMSO-de) d 1.37 (br m, 6H), 2.27 (br m, 2H), 2.80 (m, 2H), 3.07 (m, 2H), 6.98 (d, ÍH, J = 7.2 Hz) , 7.09 (t, 1H, = 7.5 Hz), 7.36 (br s, ÍH), 7.63 (dd, ÍH, J »8.5, 1.5 Hz) 7. 82 (va., 4H), 8.26 (d, ÍH, J * 7.4 Hz), 8.54 (s, 1H), 8.75 (br s, ÍH), 9.39 (s, ÍH), 12.98 (br s, 1H), 13.79 (s, ÍH). Analysis (C29H26N6 «0.7 EtOH) C, H, N.
Example 35: 3-. { [2- (5-? So-nol? N-4- l-l-nidazol-3-? L | -lfíbenzo? M dazol-4-? Lmet l] -am no.} -2-met ? l-pfopan-l-ol (a) Intermediate substance 35a C? cloprop lmet? l-. { 2- [5-? Soqu? Nolm-4-? L-l- (4-methox-bensyl) -lfl-mdazol-3-? L] -lfl-benzo? M? Dazol-4-? Lmethyl} - amine: With a procedure similar to the synthesis of 31b, alcohol 31a (512.0 mg, 1.00 mmol) was treated with methanesulfonyl and diisopropylethyl amine at a temperature of 0 ° C for 1 hour. Then aminomethylcyclopropane (712 mg, 10.0 mmol) was added, and stirring was continued at room temperature for 15 hours. After * dfel extractive treatment and column chromatography similar to substance 31b, intermediate 35a (209.3 mg, 37%) was obtained in the form of a whitish powder: Rf = 0.16 (1: 20: 300 aqueous NHOH) concentrate: ethanol: dichloromethane); XH NMR (DMSO-de) 6.-0.24 (br s, 2H), - 0.04 (br s, 2H), 0.66 (br s, ÍH), 2.30 (br s, 2H), 3.71 s, * 3H), 4.04 (brs, 2H), 5.83 (s, 2H), 6.93 (d, 2H, J = 8.7 Hz), 7.11 (m, 2H), 7.37 (d, 2H, J «8.7 Hz), 7.42 ( m, 1H), 7.67 (dd, ÍH, J = 8.7, 1.5 Hz), 7.76 (m, 2H), 7.87 (d, ÍH, J = 8.1 Hz), 8.02 (d, 1H, J = 8.7 Hz), 8.25 (dd, ÍH, J = 6.8, 1.9 Hz), 8.53 (s, ÍH), 8.68 (br s, ÍH), 9.38 (s, ÍH). Analysis (C36H32N6O »0.5H20) C, H, N. (b) Example 35-3. { [2- (5-Isoquinolin-4-yl-l-yl-indazol-3-yl) -lfl-benzoimidazol-4-ylmethyl] -amino} -2-methyl-propan-1-ol: Substance 35 was prepared in a similar manner to • example 33. The treatment of 35a (107.1 mg, 0.19 mmol) With trifluoroacetic acid / sulfuric acid in a proportion of 3: 1 supplied the open ring analog 35 (25.3 mg, 29%) in the form of white powder: Rf = »0.35 (1: 20: 100 concentrated aqueous NH4OH: ethanol: dichloromethane); l T. f , v * < NMR (CD3OD) d 0.67 (d, 3H, J = 6.8 Hz '), 1.33 (m, 1H), 1.80 * (m, 1H), 2.60 (m, ÍH), 2.75 (m, ÍH), 3.20 (m , 1H), 4.26 (S, 2H), 7.22 (m, 2H), 7.57 (d, ÍH, J = 7.7 Hz), 7.63 (dd, 1H, - 7- = 8.7, 1.7 Hz), 7.79 (m, 3H), 7.99 (d, ÍH, J * 7.5 Hz), 8.22 '(d, ÍH, J = 7.5 Hz), 8.51 (s, ÍH), 8.72 (br s, 1H), 9.29 (S, ÍH). 13C NMR (CD3OD, DEPT) d 15.0 (CH3), 35.6 (CH), 50.9 (CH2), 54.0 (CH2), 67.4 (CH2), 111.7 (CH), 123.4 (CH), 124.0 (CH), 124.3 ( CH), 125.8 (CH), 128.9 (CH), 129.3 (CH), 130.4 (CH), 132.6 (CH), 142.9 (CH), 152.6 (CH). Analysis (C28H26N6O »0.6 CH2C12» 0.4 hexanes) C, H, N. 0 Example 36: Diethyl- [2- (5-isoquinolin-4-yl-l-yl-indazol-3-yl) -1-fy-benzoimidazol-4-ylmethyl] -amine (a) Intermediate 36a-Diethyl-. { 2- [5-0 isoquinolin-4-yl-l- (4-methox? -benz? -1) -1H-indazol-3-yl] -lfl-benzoimidazol-4-ylmethyl} -amin: With a procedure similar to substance 33Ub, alcohol 31a (511.4 mg, 1.00 mmol) was treated with methanesulfonyl chloride and diisopropylethyl amine at a temperature of 5 0 ° C for 2.5 hours. Then diethylamine was added (731.4 íü * "%? *? mg, 10.0 mmol), and stirring was continued at room temperature for 25 hours. After carrying out 1 extractive treatment and column chromatography similar to substance 31b, intermediate 36av ^ s (434.6 mg, 77%) was obtained as a yellow foam: Rf = 0.22 (1: 20: 400 aqueous NH 4 OH) concentrate: ethanol: dichloromethane); XH NMR (DMSO-de) [Some peaks are doubled due to the tautomeric isomerization] 5 0.87 and 1.01 (2 brs, 6H ea 'set), 2.41 and 2.56 (2 brs, 4H as a whole), 3.71 ( s? - 10 3H), 3.89 and 3.94 (2 brs, 2H as a whole), 5.82 (s, 2H), 6.92 (d, 2H, J »8.7 Hz), 7.13 (m, 2H), 7.37 (d, 2H , J = 8.5 Hz), 7.50 (m, ÍH), 7.67 (d, ÍH, J - 8.7 Hz), 7.76 (m, 2H), 7.91 (m, ÍH), 8.01 (d, ÍH, J = 8.7 Hz), 8.25 (dd, ÍH, J - ß-. ß, * 1.9 Hz), 8.53 (s, ÍH), 8.63 and 8.77 (2 brs, ÍH as a whole) 7 9.38 (s, ÍH), 13.02 (s, ÍH). Analysis (C36H34N6? 0.4 H20) C, H, N. (b) Example 36 - Diethyl- [2- (5-isoquinolin-4 * -yl-1-yldazol-3-yl) -lfi-benzoimidazol-4-ylmethyl] -amine: Similar to example 33, the treatment of the Substance 36a (266.5 mg, 0.47 mmol) with 3: 1 trifluoroacetic acid / sulfuric acid yielded 36 (67.5 mg, 32%) in the white powder form: Rf = 0.30 (1: 20: 200 concentrated aqueous NH4OH: ethanol: dichloromethane); XH NMR (DMSO-de) [Some peaks double because of the 25 tautomeric isomerization] 5 0.94 (br m, 6H), 2.44 and 2.55 (2 ? ??? ', « br, 4H as a whole), 3.94 (brs, 2H), 7.14 (brs, 2H), - * "7.39 and 7.50 (2 brs, ÍH as a whole), 7.64 { dd, ÍH, J - ß.? * 1.5 Hz), 7.77 (m, 4H), 8.25 (d, ÍH, J = 7.4 Hz), 8.54 (faith, - | & H 1), 8.63, and 8.74 (2 brs, 1H altogether ), 9.39 (s, ÍH), * t V * ";? 12.99 (s, 1H), 13.81 (s, 1H). Analysis (C28H2eN6 »0.5 EtOH) 'C, H, N.
Example 37: Ethyl- [2- (5-isoquinolin-4-yl-l-y-indazol-3-yl) -fí-? benzoimidazol-4-ylmethyl] -amine (a) Intermediate 37a - Ethyl-. { 2-. { 5- isoquinolin-4-yl-l- (4-methoxy-benzyl) -lfl-indazol-3-yl] -l-yl-benzoimidazol-4-ylmethyl} -amine: Through a synthetic method similar to 31b, alcohol 31a (371.5 mg, 0.726 mmol) was treated with raetanesulfonyl and diisopropylethyl amine at a temperature of 0 ° C for 2.5 hours. Then the beaker of the reaction was conditioned with a digital condenser cooled with dry ice, and the ethylamine gas was condensed in the reaction solution until the volume increased in the order of 5 ral. Stirring was continued at room temperature for 15 hours. After the extractive work-up and column chromatography similar to substance 31b, intermediate substance 4a 37a (260.1 mg, 67%) was obtained in the form of a foam. pale yellow color: H NMR (DMS0-d6) d 0.84 (br s, '3H), 3.39 (br s, 2H), 3.71 (s, 3H), 4.04 (s, 2H), 5.82 (s, 2H), 6.93 (d, 2H, J = 8.7 Hz), 7.12 (m, 2H), 7.37 (d, 2H, J = 8.7 * * Hz), 7.44 (m, ÍH), 7.67 (dd, ÍH, J = 8.7, 1.5 Hz), 7.76 (m, 2H), 7.89 (m, ÍH), 8.01 (d, 1H, J = 8.7 Hz), 8.25 (dd, 1H, J = 6.6, 1.9 Hz), 8.54 (s, ÍH) , 8.67 (s, ÍH), 9.39 (s, 1H). Analysis (C34H3oNeO »0.7 H20) C, H, N. (b) Example 37-Ethyl- [2- (5-isoquinolin-4-yl-l-yl-indazol-3-yl) -lfi-benzoimidazol-4-ylmethyl] -amine: Similar to example 33, the treatment of the substance 37a (123.3 mg, 0.229 ramol) with 3: 1 trifluoroacetic acid / sulfuric acid provided 37 (21.8 mg, 23%) in the form of a whitish powder: 1 H NMR (DMSO-de) d 0.84 (br s, 3H ), 2.57 (br, 2H), 4.10 (s, 2H),. 7.13 (m, 2H), 7.46 (m, ÍH), 7.64 (dd, ÍH, J = 8.7, 1.7 Hz), "* 185 7. 80 (m, 4H), 8.26 (dd, ÍH, J = 7.2, 1.7 Hz), 8.55 (s, 1H), € .66 (s, ÍH), 9.39 (s, ÍH), 13.85 (br s, 1H ). Analysis' ^ (C2eH22N6 »0.6 EtOH« 1.0 CH2C12) C, H, N.
Example 38: Isoprop l- [2- (5- soqu? Nol n-4-? L-l-nidazol-3"l) -lfi-benzo m dazol-4- lmet l] -amine (a) Intermediate substance 38a - Isopropyl. { 2- [5-? -substituted-4-l-l- (4-methox? -benzyl) -lfl-ndazol-3-? L] -lflbenzo-m? Dazol-4-ylmethyl} -amine: With a procedure similar to substance 31b, alcohol 31a (518.0 mg, 1.01 mmol) was treated with methanesulfonyl chloride and dusopropylethyl amine at a temperature of 0 ° C for 2.5 hours. Then, isopropyl amine (597 mg, 10.1 mmol) was added and the mixture was stirred at room temperature for 24 hours. After carrying out the extractive treatment and column chromatography in a manner similar to substance 31b, intermediate 38a (417.8 mg, 75%) was obtained in the form of a yellow foam: lE NMR (DMSO-ds) d 0.77 (br s, 6H), 2.63 (br s, ÍH), 3.71 (s, 3H), 4.02 (br s, 2H), 5.82 (s, 2H), 6.93 (d, 2H, J = 8.7"Hz) # 7.11 (m, 2H), 7.37 (d, 2H, J = 8.7 Hz), 7.42 (m, 1H, 7.67 (dd, ÍH, J = 8.7, 1.5 Hz), 7.76 (m, 2H), 7.88 (d, 1H, J = 7.7 Hz), 8.02 (d, ÍH, J = 8.7 Hz), 8.25 (dd, 1H, J = 6.6, 2.1 Hz), 8.53 (s, ÍH), 8.69 (br s, 1H), 9.38 (s, ÍH) Analysis (C35H32 6? »0.7 H20) C, H, N. (b) Example 38 - Isopropyl- [2- (5-isoquinolin-4-yl-yl-mdazol-3-yl) -lfi-benzo-imidazol-4-ylmethyl] -amine: The substance of example 38 was prepared from Similar to example 33. The treatment of 38a (243.3 mg, Ó.44 mmol) with 3: 1 trifluoroacetic acid / sulfuric acid provided 38 (89.9 mg, 47%) in an off-white powder coating: 2H NMR (CD3OD ) 5 1.03 (d, 6H, J - 6.4 Hz), 2, '99 (septet, ÍH, J = 6.4 Hz), 4.27 (s, 2H), 7.23 (m, 2H), 7.57 (dd, 1H, J = 7.7, 1.1 Hz), 7.67 (dd, 1H, J - 8.7, 1.7 Hz), 7.81 (m, 3H), 8.01 (d, ÍH, J = 8.3 Hz), 8.23 (d, ÍH, J * 7.5 Hz ), 8.51 (s, 1H), 8.71 (br s, ÍH), 9.30 (s, 1H).
Example 39: tert -Butyl- [2- (5-? Soqu? Nolm-4-yl-l-yldazol; -3-, yl) -f-benzoimidazol-4-ylmethyl] -amine < * ' (a) Intermediate substance 39a-tert-Butll-t2- [5 ~ -. isoquinolin-4-yl-l- (4-methoxy-benzyl) -l-fy-indazol-3-yl] -lH-benzoimidazol-4-yl-methyl} -amine: By a method analogous to 31b, alcohol 31a (623.2 mg, 1.22 mmol) was treated with t-methanesulfonyl chloride and diisopropylethyl amine at a temperature of 0 ° C for 1 hour. Then tert-Butylamine f890"* mg, 12.2 mmol) was added and stirring was continued at room temperature for 20 hours.
Extractive treatment and column chromatography of man V similar to substance 31b, intermediate 39a (299.7 mg, 43%) was obtained in the form of a yellow foam: XH NMR (CD30D) 5 1.01 (s, 9H) , 3.76 (s, 31), 4.11 (s, 2H), 5.78 (s, 2H), 6.91 (d, 2H, J = 8.7 Hz), 7.19 (m, 2H), 7.36 (d, 2H, J = 8.7 Hz), 7.50 (dd, ÍH, J = 7.9,; -1.1Hz), 7.62 (dd, ÍH, J = 8.7, 1.7 Hz), 7.77 (m, 3H), 7.95 (d, ÍH, J * 7.9 Hz ), 8.22 (dd, ÍH, J = 7.0, 1.7 Hz), 8.48 (s, ÍH), 8.74 (s, 1H), 9.29 (s, ÍH). Analysis (C36H34NeO »0.3 * • 'H20) C, H, N. (b) Example 39 - tert -Butyl- [2- (5-isoquinolin-4-yl-lfl-indazol-3-yl) -lfi- benzo-imidazol-4-yl-met? l] -amine: A solution of 39b (103.7 mg, 0.183 mmol), trifluoromethanesulfonic acid (0.48 ml) and trifluoroacetic acid (1.6 ml), was stirred at room temperature for 17 hours and then at 100 ° C for 1.5 hours.The solution was added by faith '* * S By a method analogous to 31b, alcohol 3j¿ (572., O mg, 1.12 ramol) was treated with methanesulfonyl chloride and dusopropylethyl amine at an a cc temperature, 0 ° C for 1 hour. Then 1-idazole (761 mg, 11.2 mmol) was added and stirring continued at room temperature for 24 hours. After performing the extractive treatment and column chromatography in a manner similar to substance 31b, intermediate 40a (269.1 mg, 43%> as a white powder: XH NMR (CD3OD) d 3.77 (s, 3H), 5.58 (s, 2H), 5.79 (s, 2H), 6.73 (br s, ÍH), 6.91 (d, 2H, J = 8.8 Hz), 7.07 (d, ÍH, J = 7.4 Hz), 7.23 (m, 2H), ^ 7.36 (d, 2H, J = 8.8 Hz), 7.53-7.83 (m, 6H), 8.03 (d, ÍH ,, = 7.9 Hz), 8.22 (d, 1H, J * 7.9 Hz ), 8.51 (s, ÍH), 8.73 (bg _ '^ s, ÍH), 9.28 (s, ÍH), Analysis (C35H27N70) C, H, N. (b) Example 40-4- [3- (4-Im? dazol-1-yl-metrA-l-2-benzyl or 2-ol-2-? l) -lfl-nda-2-yl-yl] -isoquinolma: A solution of 40a (152.0 mg, 0.271 mmol), trifluoromethanesulfonic addict (0.271 ml) and trifluoroacetic acid (2.71 ml) was stirred at a temperature of 60 ° C for 1 hour. The solution was added dropwise to a mixture which was kept under rapid stirring of concentrated aqueous NH40H. - (10 ml), water (10 ml), THF (10 ml) and ethyl acetate (2fr »? Ml). Extraction and purification similar to example 33, gave the crude compound 40 in the form of solid pink (24.9 mg), which still had impurities * "V ,, t < 190 in the 1H NMR spectrum. Trituration of acetonitrile gave pure compound 40 (11.0 mg, 9%) as a pink powder: H NMR (CD3OD) d 5.59 (s, 2H), 6.74 (br s, 1H), 7.08 (d, 1H, J = 7.4 Hz), 7.25 (m, 2H), 7.55-7.85 (m, 6H), 8.07 (d, ÍH, J = 7.9 Hz), 8.24 (d, ÍH, J = 7.5 Hz), 8.54 ( s, ÍH), 8.72 (br s, ÍH), 9.30 (s, ÍH). HRMS calculated for C27H2oN- 442.1780 (MH +), found 442.1794.
Example 41 5- (3-Met? L-p nd? N-4-? L) -3- (E) -est? Ril-lH-mdazole (a) Intermediate 41a-5- (3-Met? l-pyridin-4-yl) -3- ((E) -est ril) -1- (2-tr? met? ls? lan? l-etox ? met l) -lfí-indazol: Intermediate 16a (300 mg, 0.63 mmol), 4-bromo-3-met? lp? r? dma (See Baliki et al., Gazz. Chim. Ital. 124, 9 , 1994, 385-386) (112 mg, 0.65 mmol) and carbonate v4 191 Sodium (140 mg, 1.3 mmol) was stirred in DME (6 ml) / H20 (1 * ml) in a beaker purged with argon. "Tetrakis (tp-phenylphosphine) palladium (0) (60 mg, 0.05 mmol) was added and the reaction was stirred at reflux temperature for 24 hours.The solution was diluted with ethyl acetate, washed with H20 and brine, dried over Na2SO4 and concentrated in vacuo Purification by chromatography on silica gel (20% ethyl acetate / hexanes) gave 234 mg (84%) of intermediate 41a as a clear oil ? E NMR (300 MHz, CDC13) d 8.56 (s, 1HX, 8.52 (d, ÍH, J = 7.8 Hz), 7.95 (s, ÍH), 7.24-7.67 (m, 10H), 5.78 (s, 2H ), 3.64 (t, 2H, J = 8.1Hz, 2.33 (s, 3H), 0.94 (t, 2H, J = 8.1Hz), -0.04 (s, 9H) Analysis (C2H3iN3OSi 0.2 H20) C, H, N. (b) Example 41-5- (3-Met? l-pyridin-4-yl) -3- (E) -styryl-mdazole: Intermediate 41a (218 mg, 0.49 mmol) was stirred in a mixture of ethylenediamine (0.34 m, 4.9 mmol) and TBAF (1 M in THF, 2.5 m, 2.5 mmol) at a temperature of 72 ° C for 20 hours. The solution was diluted with ethyl acetate, washed with saturated NaHCO 3 and brine, dried (Na 2 SO 4) and concentrated in vacuo. Purification by chromatography on silica gel (1: 1: 1 ethyl acetate / THF / hexanes) gave 122 mg (79%) of the title compound as a white solid.1H NMR (300 MHz, DMSO-de) 5 13.29 (s, 1H), 8.52 (s, 1H), 8.46 (d, 1H, J »'*" < 4.8 Hz), 8.22 (s, ÍH), 7.55-7.73 (m , 5H), 7.26-7.44 (m, 5H), 2.31 (s, 3H) Analysis (C2iH? 7N3) C, H, N. Ms' (ES) [m + HJ / z calculated 312, found 312; mH] / z calculated 310, found 310.
Example 42: 5- (4-Chloro-p? Ridin-3-yl) -3- (E) -est? Ryl-l-yl-indazole (a) Intermediate 42a-5- (4-Chloro-pyr-din-3-yl) -3- ((E) -estipl) -1- (2-tnmethylsilanyl-ethoxymethyl) -lfl-mdazole: The compound of the title was prepared in a 73% yield from intermediate 16a and 4-chloro-3-iodo-pyridine (See Cho et al., Heterocycles, 43, 8, 1996, 1641-1652) analogous to intermediate 41a. ^ NMR (300 MHz, CDC13) 5 8.67 (s, ÍH), 8.52 (d, ÍH, J = 7.8 Hz), 8.08 (s, 1H), 7.26-7.70 (m, 10H), 5.79 (s, 2H) 3.64 '193 ~ s • - (t, 2H, J «8.1Hz), 0.94 (t, 2H, J = 8.1Hz), * 0.03 (s, 9H). ^ t Analysis (C26H28ClN3OSi »0.3H20) C, H, N. 'X i (b) Example 42- 5- (4-Chloro-pyridin-3? l) -3- (E) -T%? f. "" estiril-lfí-indazol: it- The title compound was prepared in q? - * 66% yield by means of the deprotection SEM of the V. "" * "intermediate 42a in a method analogous to the example -. '* < 41. ^ NMR (300 MHz, DMSO-de) d 13.30 (s, ÍH), 8.70 (s, ÍH), I 8.56 (d, 1H, J = 5.4 Hz), 8.31 (s, ÍH), 7.63-7.73 (m, 4H), * V 0 7.57 (d, 2H, J = 4.2 Hz), 7.50 (dd, ÍH, J - 8.4, 1.2 Hz), 7.26-7.40 (m, 3H). Analysis (C2oH14ClN3 • 0.05H20) C, H, N. '' - # MS (ES) [m + H] / z calculated 332/334, found 332/334; [m-H] / z *, calculated 330/332, found 330/332.
Example t 43: 5- (4-Methyl-pyrid? N-3-yl) -3- (E) -styryl-1 ^ -inda2 l - (a) Intermediate 43a-5- (4-Met? lp? pdin-3-yl) -3- ((E) -estipl) -1- (2-tpmethyls? lanil-etox? met? l) -lff Inda2?: The title compound was prepared in a 90% yield from intermediate 16a and 3-bromo-4-met? lp? r? dma in a manner similar to the procedure used for intermediate 41a. 1 H NMR (300 MHz, * CDCl 3) d 8.54 (s, ÍH), 8.50 (d, ÍH, J * 7.8 Hz), 7.95 (s, 1H), 7.23-7.67 (m, 10H), 5.78 (s, 2H ), 3.64 (t, 2H, J = 8.1Hz), 2.33 (s, 3H), 0.94 (t, 2H, J = 8.1Hz), -0.04 (s, 9H). Analysis (C27H3? N3OS?) C, H, N. (b) Example 43-5 (4-Met? lp? r? din-3-? l) -3- (E) -st? pl-lfl-mda2ol: The title compound was prepared in an -end? 48% by means of the SEM deprotection of intermediate 43a in a method analogous to example 41. H NMR (300 MHz, DMSO-d6) d 13.26 (s, ÍH), 8.47 (s, 1H), 8.44 (d , 1H, J = 4.8 Hz), 8.20 (s, ÍH), 7.71 (d, 2H, J = 7.2 Hz), 7.55-7.64 (m, 3H), 7.26-7.42 (m, 5H), 2.31 (s, 3H). * Analysis (C2? H? 7N3 • 0.13 H20) C, H, N. MS (ES) [ta-H] / z calculated 312, found 312; [m-H] / z calculated 310, found 310. to ^ -ispigtt omina J # (a) Intermediate 44a-4-Bromo-5-fluoro-isoquinoline: 5-Am? no-4-bromo-? soqu? nolma (See Gordon et al., J. Heterocycl. Chem., 4, 1967, 410 -411) (1.86 g, 8.34 mmol) was stirred in 48% fluoroboric acid (15 ml) / EtOH (15 ml) until completely dissolved. The solution was cooled to a temperature of 0 ° C and sodium nitrite (660 mg, 9.59 mmol) in H20 (1 ral) was added dropwise. The solution was diluted with Et20 (30 ml) and the tan-colored diazomene fluoroborate salt was collected by filtration and dried in vacuo. The solid was placed in a beaker and heated slowly over a flame to expel the nitrogen. The dark brown residue was diluted with 10% NAOH and extracted with chloroform. The organic compounds were washed with brine, dried over MgSO and they concentrated in vacuum. Purification by chromatography on silica gel (40% to 50% ethyl acetate / hexanes), gave 798 mg (42%) of 4-bromo-5-fluoro-isoquinoline as a white solid XH NMR (300 MHz, CDCls) 9.36 (d, 1H, J-2.4 Hz), 8.74 (s, ÍH) 8.07-8.11 (m, 1H), 7.70-7.80 (m, 2H). Analysis (C9H5BrFN) C, H, N. (b) Intermediate 44b-5-Fluoro-4- [3- (D-styryl) -1- (2-trimethylsilanyl-ethoxymethyl) -1-fy-indazol-B-yl] -isoquinoline: The title compound was prepared in an 83% yield from intermediate 16a and 4-bromo-5-fluoro-isoquinoline similar to intermediate 41a. XH NMR (300 MHz, CDC13) d 9.32 (d, ÍH, J - 1.8 Hz), 8.52 (s, ÍH), 8.07 (s, ÍH), 7.91 (dd, ÍH, J = 8.1, 0.9 Hz), 7.26 -7.66 (m, 11H), 5.80 (s, 2H), 3.67 (t, 2H, J = 8.1Hz), 0.95 (t, 2H, J - 8.1Hz), -0.03 (s, 9H). Analysis (C30H30FN3OSÍ • 0.2 H20) C, H, N. (c) Example 44- 5-Fluoro-4- [3- ((E) -styryl) -lfi- "indazol-5-yl] -isoquinoline: The title compound was prepared in an 83% yield by SEM deprotection of intermediate 44b in a manner analogous to example 41. XH NMR (300 MHz, DMSO-de) d 13.26 (s, ÍH), 9.44 (d, 1H, J * = 1.8 Hz), 8.47 (s) , ÍH), 8.29 (s, ÍH), 8.12 (d, 1H, J «7.2 ' Hz), 7.44-7.78 (m, 8H), 7.35 (t, 2H, J = 7.2 Hz), 7.24 (t, ÍH, J = 7.2 Hz). Analysis (C24H16 N3 • 0.6H20) C, H, N.-MS (ES) [m + H] / z calculated 366, found 366; [m-H] / z calculated, ^ 364, found 364.
E 45 4 - [3- ((E) -styryl) -lfi- adazol-5-yl - isoquinol n-8-jlam na (a) Intermediate 45a-4 [3- ((E) -styryl) -l- (2-tr? met? ls? lan? -ethox? met? l) -lfl-? ndazol-5-? ] -? soqumol? n-8-? lam? na: The title compound was prepared in an 82% yield from the intermediate substance read and 8-am? no-4-bromo-? soqumolma (See Elpern and col., J. Amer, Chem. Soc., 68, 1946, 1436) similarly to the procedure used for intermediate 41a. 1 H NMR (300 MHz, CDCl 3) d 9.36 (d, HH, J = 0.6 Hz), 8.53 (s, 1H), 8.13 (s, HH) 7.26-7.72 (m, 11H), 6.86 (dd, HH, J = 7 «5, 'i * s? T "* 5 * 198 kr. 0.6 Hz, 5.81 (s, 2H), 51 (s, 2H), 3.66 (t, 2H, J - 8.1Hz), 0.96 (t, 2H, J 8.1 Hz), -0.03 (s, 9H). (b) Example 45- 4- [3- ((E) -styryl) -lfl-mdaz6r-5- ^ A. I '* il] -? soqu? nolin-8-ylamine: X The title compound was prepared in a 70% yield by means of the SEM deprotection of the. intermediate 45a analogously to the procedure used for example 41. XH NMR (300 MHz, DMSO-dg) d O 13.30 (s, ÍH), 9.50 (s, ÍH), 8.36 (s, 1H), 8.26 (s) ,? .H. T - 7.24-7.71 (m, 10H), 6.91 (d, ÍH, J = 7.8 Hz), 6.77 (t, ÍH, J = 7.8 Hz), 6.33 (s, 2H) .Analysis (C24H18N4"0.45 H20) C, H N. MS (ES) [m + H] / z calculated 363, found 363.
Example 46 5- (4-Chloro-5-ethyl-pyridin-3-yl) -3 - (»B) - styryl-1-yl-indazole [Dioxane - Dioxane] 5 ' ml) at a temperature of -20 C. After 10 minutes, the solution was cooled to -78 ° C. 4-4 Chloro-3-iodopyridone (500 mg, 2.09 mmol) in THF (3 mL) was added dropwise, and the reaction was stirred for 30 minutes. Iodoethane (0.2 m, 2.5 mmol) was added, and the reaction was stirred for 1 hour at a temperature of -78 ° C, then for 1 hour while heating to 0 ° C. The reaction was warmed with saturated NH 4 Cl, basified with saturated NaHCO 3 and extracted with and ethyl acetate. The organic compounds were washed with brine, dried over Na 2 SO 4 and concentrated in vacuo. Purification by chromatography on silica gel (20% ethyl acetate / hexanes) provided 429 mg (77%) of 4-Chloro-3-et? L-5-iodo-p? R? Dma in the form of a waxy solid white color. ^ NMR (300 MHz, CDC13) d 8.78 (s, ÍH), 8.33 (s, ÍH), 2.83 (q, 2H, J - 7.5 Hz), 1.26 (t, 3H, J = 7.5 Hz) . Analysis (C7H7CIIN) C, H, N. r (b) Intermediate substance 46b-5- (4-Chloro-5-et? lp? r? dm-3-? l) -3- ((E) -estipl) -1- (2-trimethylsilanyl-ethoxymethyl) -lfí-mdazol: .2 * ?. * -fc.
The title compound was prepared in a 69% yield from intermediate 16a and 4-chloro-3-et? L-5-iodo-p? R? Dma in a manner similar to the procedure used for intermediate 41a . XH NMR (300 MHz, CDC13) d 8.49 (d, 2H, J- 3.3 Hz), 8.06 (s, 1H) 7.26-7.69 (m, 9H), 5.79 (s, 2H), 3.65 (t, 2H, J = 8.1Hz), 2.88 (q, 2H, J = 7.5 Hz), 1.35 (t, 3H, J = 7.5 Hz), 0.95 (t, 2H, J = 8.1Hz), -0.03 (s, 9H). Analysis (C28H32ClN3OSi) C, H, N. (c) Example 46-5- (4-Chloro-5-ethyl-pyridin-3-yl) -3- (E) -styryl-1-yl-indazole: The title compound was prepared in an 80% yield by of the SEM deprotection of the intermediate substance 46b analogously to the procedure used for example 41. XH NMR (300 MHz, DMSO-de) d 13. * 25 (s, ÍH), 8.55 (s, ÍH), 8.50 (s, ÍH), 8.27 (s, 1H), 7.55-7.72 (m, 5H), 7.26-7.48 (m, 4H), 2.83 (q, 2H, J »7.5 Hz), 1.26 (t, 2H, J * 7.5 Hz). Analysis (C22H18N3C1 • 0.3H20) C, H, N. MS (ES) [m + H? / Z calculated 360, found 360.
Example 47: 3- [3- (1-Benzoimdazol-2-yl) -lfí-in < jtazol-5-yl-2-methoxy-phenol 47a 47 [Benzene = Benzene Reflux = Reflux] (a) Intermediate 47a-3- (1-benzoamidazol-2-? l) -5-. { 2-methoxy-3-. { 2- (2-trimethylsilanyl-ethoxymethyl) -ethoxy] -phenyl} -l- (2-tr? methylsilanyl-ethoxymethyl) -Ifl-mdazole: The title compound was prepared in a yield of 92% from the intermediate 7c 'and 2-methox? -3- [2- (2-methyl-ethoxy) -ethoxy-boronic acid (See: Kania, Braganza, et al., Patent application "Compounds and Pharmaceutical Compositions for Inhibition of Protein Kinases, and Methods for Their Use", p. 52, line 10 to page 53, line 26, and page 59, line 16 to page 60, line 4, US Provisional Serial number 60 / 142,130, filed on July 2, 1999, which is incorporated herein in its entirety as a reference.), similar to the '** V? Fk 95% yield from intermediate 7cf and intermediate 48b similarly to the procedure used for intermediate 7d .. 1H NMR (300 MHz, CDC13) d 9.90 (s, ÍH), 8.96 (d, 1H, J »0.9 Hz), 7.84-7.90 (m, 2H), 7.69 (d, ÍH, J * 8.7 Hz), 7.48-7.53 $ (m, 2H), 7.23-7.33 (m, 5H), 6.82 (d, ÍH, J «3.3 Hz), 5.8 ^? (s, 2H), 3.67 (t, 2H, J = 8.1Hz), 0.96 (s, 9H), 0.94 (t, 2H, J = 8.1Hz), 0.65 (s, 6H), - 0.03 (s, 9H) ). Analysis (d) Example 48-3- (1-Benzo? m? dazol-2-? l) -5- (1-ylfol-4? l) -lfl-mdazole: The title compound was prepared in a yield of 79% by means of the deprotection SEM, TBDMS of intermediate 48c analogously to the procedure used for example 41. 1 H NMR (300 MHz, DMSO-de) d 13.66 (s, ÍH), 12.97 (s, 1H) , 11.25 (s, ÍH), 8.78 (s, ÍH), 7.68-7.81 (m, 3H), 7.51 (d, 1H, J = 7.2 Hz), 7.42- (91%) of the title compound in the form of a clear oil. XH NMR (300 MHz, CDC13) d 7.11-7.20 (m, 1H), 6.74-6.89 (m, 2H), 5.20 (s, 2H), 3.77-3.83 (m, 2H), 0.93- 4. 0.99 (m, 2H), 0.01 (s, 9H). X (b) Intermediate 49b-3-fl # ~ Benzo? m? dazol-2-? l) -5. { 2,6-difluoro-3- [2- (2-tpraet? Ls? Lanyl-ethoxy) -ethoxy] -phenyl} -1- (2-tr? Met? L? L? -ethoxymethyl) -lfi-indazole: Intermediate 49a (1.4 g, 5.38 mmol) was stirred in dry THF (16 ml) under argon at a temperature of - 78 ° C. N-Butyllithium (2.5 M in hexanes, 2.32 m, 5.8 mmol) was added dropwise, and the reaction was stirred for 20 minutes. Then, the solution was transferred by means of a cannula to a beaker of dry zinc chloride in argon at room temperature. After 30 minutes, intermediate 7c '(320 mg, 0.65 mmol) and tetrakis (tp-phenylphosphine) palladium (0) (60 mg, 0.05 mmol) were added, and the reaction was stirred at room temperature for 2 hours. it was diluted with ether and washed with H20, saturated NaHCOa, and brine. The organics were separated over Na 2 SO 4 and concentrated in vacuo. Purification by *, chromatography on silica gel (20% to 30% Et20 / hexanes) gave 372 mg (92%) of the title compound as a white solid. XH NMR (300 MHz, CDCl 3) d 9.89 (s, ÍH), 8.80 (s, ÍH), 7.86-7.89 (m, ÍH), 7.70 (dd, ÍH, J = 8. 7, 0.9 Hz), 7.58 (dd, ÍH, J = 8.7, 1.2 Hz), 7.49-7.53 (m, 1H), 7.17-7.31 (m, 3H), 6.90-6.97 (m, ÍH), 5.82 (s, 2H), 5. 28 (s, 2H), 3.86 (t, 2H, J = 8.4 Hz), 3.67 (t, 2H, J * 8. 1Hz), 0.92-1.04 (m, 4H), 0.02 (s, 9H), -0.02 (s, 9H). - * Analysis (C32H4oF2N4? 3Si2 «0.25H20) C, H, N." • * (s) Example 49 - 3- [-3- (lfl-Ben2? Imidazol-2-yl) - lfí-inda2 l-5 -yl] -2,4-difluoro-phenol: The title compound was prepared in a 70% yield by means of the SEM deprotection of intermediate 49b analogously to the procedure used for example 41, XH NMR (300 MHz , DMSO-de) d 13.75 (s, ÍH), 13.00 (s, ÍH), 9.88 (s, ÍH), 8.56 (s, 1H), 7.70-7.78 (m, 2H), 7.48-7.53 (m, 2H ), 7.17-7.25 (m, 2H), 6.99-7.05 (m, 2H). Analysis (C20H? 2FN4? • 0.33H20) C, H, N. MS (ES) [m + H] / z calculated 363, found 363; [m-H} / z calculated 361, found 361.
Example 50: 4 ^ [-3- (1-Benzyl-imidazol-2-yl) -Ifl-indazol-S -yl] -3,5-difluoro-phenol and $ i example 51; 2- [-3- (lfl-Benzoyrazol-2-yl-2-yl) -lfl-inda2? L-5-yl]; B, 5-djf luoro-f enol 6. 40-6.48 (m, ÍH), 5.18 (s, 2H), 3.70-3.76 (m, 2H), 0. St2- 0.98 (m, 2H >, 0.01 (s, 9H). (b) Intermediate ring of 50b and 50c-3-fifi-Benzoyl-tidazol-2-yl) -5. { 2,6-difluoro-4- [2- (2-trimethylsilartyl-ethoxy-ethoxy] -phenyl] -1- (2-trimethylsilanylethyloxymethyl) -Ifl-Indazole and 3-fi- Benzoimidazol-2-yl) -5 . { 2,4-difluoro-6- [2- (2-trimethylsilanyl-ethoxy-ethoxy] -phenyl] -l- (2-trimethylsilanyl-ethoxymethyl) -lfl-indazole: The title compounds were prepared in a yield of 52% as an inseparable mixture from intermediate 50a similar to the procedure used for intermediate 49b, 1 H NMR (300 MHz, CDC13) d 9.89 (s, HH), 8.44-8.75 (m, 1H), 7.83- 7.93 (m, ÍH), 7.45-7.69 (m, 3H), 7.26-7.39 (m, 2.5H), 6.58- 6.88 (m, 1.5H), 5.81 (s, ÍH), 5.80 (s) , ÍH), 5.26 (s, lfí), 5.13 (s, ÍH), 3.57-3.82 (m, 4H), 0.86-1.04 (m, 4H), - 0.06-0.02 (m, 18H). (c) Example 50-4- [-3- (1-Benzoy-Nidazol-2-yl) -lfl-mdazol-5-yl] -3,5-difluoro-phenol: The title compound was prepared in a yield 36% by means of the SEM deprotection of the mixture of intermediates 50b and 50c in a manner analogous to the procedure used for the 41th model. XH NMR (300 MHz, DMSO-de) d 13.73 (s, ÍH), 13.01 (s) , 1H), 10.50 (s, ÍH) / ** '• 8. 50 (s, 1H), 7.70-7.74 (m, 2H), 7.43-7.52 (m, 2H), 7.15-7.25 (m, 2H), 6.62 (dd, 2H, J = 13.8, 1.5 Hz). Analysis (C2oHX2FN40 «0.7 H20) C, H, N. MS (ES) [m + H] / z calculated 363, / &'; Found 363; [m-H] / z calculated 361, found 361, (d) Example 51-2- [-3- (1-Benzoimid zol -2-il) - < Í lfí-indazol-5-? L] -3, 5dif luoro-f enol: The title compound was prepared in * a yield of 40% by means of the deprotection SEM of the "r intermediate mixture 50b and 50c in an analogous manner. al procedure used for example 41. XH NMR (300 Mfiz, DMSO-de) d 13.65 (s, ÍH), 12.98 (s, 1H), 10.39 (s, ÍH), 8.47 - ^ (s, 1H), 7.66- 7.72 (m, 2H), 7.50 (d, 1H, J - 7.2 Hz), 7.40 (d, 1H, J = 8.4 Hz), 7.14-7.24 (m, 2H), ß.73-6.80 (m, 1H) , 6.64 (d, ÍH, J = 10.5 Hz). Analysis (C20H12FN4? • 0.9H20) C, * H, N. MS (ES) [m + H] / z calculated 363, found 363; [m-IJ / z calculated 361, found 361.
Example 52: 3- (1-Benzoim dazol-2-yl) -5- (4-clpro-pyridin-3-yl) -lfi-Indazole * [Dioxane = 0 (a) Intermediate 52a-3- (1-Benzo-2-trifluoro-2?) -5- (4-chloro-p? R? Din-3-yl) -1- (2-t-imethylsilanyl-ethoxymethyl) -lfl-mdazole: The title compound was prepared in an 88% yield from intermediate 25a 5 and 4-chloro-3-iodo-p? r? d? na in a manner similar to the procedure used for the substance intermediate 41a. XH NMR (300 MHz, CDC13) d 9.96 (s, ÍH), 8.77 (s, ÍH), 8.72 (s, ÍH), 8.53 (d, ÍH, J - 5.4 Hz), 7.85-7.89 (m, ÍH) , 7.72 (dd, ÍH, J = 8.7, $ 0.9 Hz), 7.61 (dd, ÍH, J = 8.7, 1.5 Hz), 7.50-7.53 (m, 1H), 7.47 (d, 1H, J - 5.4 Hz), 7.28-7.35 (m, 2H), 5.84 (s, 2H) 3.65 (t, 2H, J = 8.1Hz), 0.95 (t, 2H, J - 8.1Hz), -0.03 (s, -, 9H). (b) E jmplo 52-3- (lfí-Benzo? m? dazol -2-il) -5- 5 '< 4 -Cloro-p? R? D? N-3 -? L) - lfí-mdazol: The title compound was prepared in a 54% yield by means of the deprotection SEM of the intermediate substance rfi 52a analogously to the procedure used for example 41. XH NMR (300 MHz, DMSO-de) 13.81 (s, ), 13.02 (s, 1H), 8.70 (s, ÍH), 8.56-8.60 (m, 2H), 8.22 (s, ÍH), 7.55-7.80 (m, 5H), 7.20 (d, 5H, J = 3 , 6 Hz). Analysis (C22H18CI 3 • 0.5H20) C, H, N. MS (ES) [m + HJ / z calculated 346, found 346.
Example 53: 5- [3- (1-Benzoimidazol-2-yl) -lfl-indazoX-S-yl] -4-methyl- [3, 4 '] bipyridinyl [Dioxane = Dioxane] (a) Intermediate 53a 5- (Bromine * "4-5 methyl- [3, '] bipyridinyl: faith 214 3, 5-D? Bromo-4-met? Lp? Pd? Na (2.21 q, 8.0 mmol), * 4-p? Rid? Lboron? Co (1.08 g, 8.8 mmol) and phosphate ** > ,you*. potassium (2.8 g, 13.2 mmol) was stirred in DMA (50 ml) / H0 (6 ml) in a beaker purged with argon. HE Tetrakis (tpfenilfosfma) palladium (0) (812 mg, 0.7 mmol) and and ** the reaction was stirred at a temperature of 92 ° C under argon; for 16 hours. The solution was concentrated in vacuo and the residue was dissolved in ethyl acetate. The organic compounds? They were washed with H20 and brine, dried (Na2SO4) and concentrated in vacuo. Purification by chromatography on silica gel (40% to 50% ethyl acetate / hexanes) yielded 1.14 g (60%) of the intermediate substance * * 53a as a white solid. XH M - (300 MHz, CDC13) d 8.73 (dd, 2H, J = 4.5, 1.5 Hz), 8.72 (s, "ÍH), 8.32 (s, ÍH), 7.25 (dd, 2H, J - 4.5, 1.5 Hz), 2.35 (S, 3H), Analysis (C ??H9BrN2) C, H, N. (b) Intermediate substance 53b-5- [3- (l-Benzoim? dazol-2-? l) -1- (2-tnmet? ls? lan? l-ethoxy? tnet? l) -lfl-ndazole- 5-? L] -4-met? L- [3, 4 '] b? P? Pd? N? L: The title compound was prepared in a 64% yield from intermediate 25a X and intermediate 53a in a manner similar to the procedure used for intermediate 41a. 1H NMR (300 MHz, CDC13) d 10.21 (s, ÍH), 8.70-8.76 (m, 3H), 8.61 (s, ÍH), 8.46 (s, ÍH), 7.85 8.7, 0.9 Hz), 7.47-7.53 (m, 2H), 3.64 (t, 2H, J = 8.1Hz), 2.19 (s, 3H), 0.94 (t, 2H, J *. 8.1Hz), -0.04 (s, 9H). (c) Example 53-5- [3-l-Benzoimidazol-2? l) -lfl-mdazol-5? l] -4-met? l [3,4 '] b? p? r? d? ml: The title compound was prepared in a 71% yield by means of SEM deprotection dβ intermediate 53b analogously to the procedure used for example 41. XE NMR (300 MHz, DMSO-dβ) d 13.79 (s) , ÍH), 13.02 (s, ÍH), 8.71 (d, 2H, J = 4.8 Hz), 8.55 (s, ÍH), 8.51 (s, ÍH), 8.47 (s, ÍH), 7.78 (d, ÍH, J - $ .7 Hz), 7.52-7.58 (m, 5H), 7.18-7.21 (m, 2H), 2.17 (s, 33.} .. Analysis (C25Hi8N6 • 0.75 H20) C, H, N. MS (ES) [m + H] / z-calcified 403, found 403; [mH] / z calculated 401, found 401.
Example 54: 5- [3- (l-Benzo-m-dazol-2-yl-l-l-ndazol-5-yl] -l, -2, 3, 4, 4a-8a-hexahydro- [l, 7] ] naft? r dina < * * * For 1 hour at a temperature of -78 ° C and then last 1 hour while heating it to 0 ° C. ! * > & , reaction was warmed with saturated NHC1, basified with saturated NaHCOg and extracted with ethyl acetate. The organic compounds were washed with brine, dried over Na 2 SO 4 and concentrated in vacuo. Purification by chromatography on silica gel (15% MeOH / CHCl3) provided 2.72 g (54%) of the intermediate 54a in the form of a light brown oil. XH NMR (300 MHz, CDCl 3) d 8.55 0 (d, 2H), 2.72-3.05 (m, 6H), 1.70-1.77 (m, 2H). (b) Intermediate substance 54b - 2- Jí acid > * Trimethylalanyl-ethanesulfonic acid [3- (3,5-dibromo-pi idin-> 4-yl) -propyl] -amide: "i. Intermediate 54a (2.7 g, 9.2 mmol) was stirred with dry triethylamine (1.92 m, 13.8 ramol) in DMF (20 ml) at a temperature of 0 ° C. 2-Tpmethylsilyanil-ethanesulfonyl chloride, SES-CI, was slowly added (See einreb and COl., Tet.Lett., 27, 19, 1986, 2099-2102) (1.9 g, 9.5 mmol), and the reaction was stirred for 1.5 hours at a temperature 0 of 0 ° C. The reaction was diluted with H20 and extracted with ether. The organic compounds were washed with brine, dried over Na 2 SO and concentrated in vacuo. Purification by chromatography on silica gel (33% ethyl acetate / hexanes) provided 2.37 g (56%) of the intermediate 54b as a colored solid White. lE NMR (300 MHz, CDC13) d 8.58 (s, 2H), 4.36 (t, 1H J «6.3 * Hz), 3.26 (q, 2H, J = 6.3 Hz), 2.93-3.06 (m, 4H),« 1.81-1.89 (m, 2H), 1.00-1.07 (m, 2H), 0.07 (s, 9H). Analysis (C13H22Br2N202SS?) C, H, N, S. (c) Intermediate 54c-5-Bromo-l- (2-trimethylsilanyl-ethanesulfonyl) -1,2,3,4-tetrahydro- [1/7] naphthyridine: Intermediate 54b (860 mg, 1.88 mmol) and Potassium carbonate (390 mg, 2.82 mmol) was stirred in dry toluene (15 ml) in a beaker purged with argon. Tetrakis (triphenylphosphine) palladium (0) (218 mg, 0.19 mmol) was added and the reaction was stirred under argon at 102 ° C for 48 hours. The reaction was diluted with ethyl acetate and washed with brine, dried (Na2SO4) and concentrated in vacuo. Purification by chromatography on silica gel (25% ethyl acetate / hexanes) gave 372 mg (52%) of intermediate 54c in the form of a white waxy solid. XH NMR (300 MHz, CDCl 3) d 8.70 (s, ÍH), 8.42 (s, ÍH), 3.72-3.76 (m, 2H), 3.07-3.14 (m, 2H), 2.83 (t, 2H, J »6.9 Hz), 2.07-2.12 (m, 2H), 1.04-1.11 (m, ~ 2H), 0.05 (s, 9H). Analysis (C13H2iBrN2? 2SSÍ) C, H, N, S. (d) Intermediate 54d-5- [3- (1-Benzo [mu] midazol-2-yl) -1- (2-tr? methylsilyl-ethoxymethyl) -Ifl- ? > * indazol-5-yl] -1- (2-trimethylsilanyl-ethand ulfo? il) -l, ^ 3 and 4, 4a, 8a-hexahydro- [1,7] naphthyridine: The title compound was prepared in T-yield of the 51% from the intermediate substance - '/ and from the intermediate 54c similar to the procedure used for intermediate 1 & 1H: '.-: NMR (300 MHz, CDC13) d 10.36 (s, ÍH), 8.88 (s, 1H), 8.6; (t,.;;,. ÍH, J »0.9 Hz), 8.29 (s, ÍH), 3.82-3.86 (m, '1H), 7.69 (dd,; lH, J * 8.7, 0.9 Hz), 7.50 -7.52 (m, ÍH), 7.41 (dd, ÍH,; J = "y 10 8.7, 1.5 Hz), 7.26-7.33 (m, 2H), 5.83 (s, 2H), 3.80 (t, 2H ,; - J = 5.7 Hz), 3.63 (t, 2H, J = 8.1Hz), 3.13-3.20 (m, 2H), \ 2.72 (t, 2H, J = 6.6 Hz), 1.93-1.99 (m, 2H), 1.10-1.16 (m, r '.2H), 0.94 (t, 2H, J - 8.1Hz), 0.09 (s, 9H), - 0.O5 (s, 9H) .. P. (e) Example 54 -5- [3- (1-Benzoimidazol-2-yl) -l, 15-yl-indazol-5-yl] -1,2,3,4,4a, 8a-hexahydro- [1, 7] Naphthyridine: The title compound was prepared in a 64% yield by deprotection SEM and SES '"of intermediate 54d analogously to 20-procedure used for Example 41. XH NMR (300 JMHz, DMSO-de ) d 13.71 (s, ÍH), 13.00 (s, ÍH), 8.41 (s, .H),; 7.82 (s, ÍH), 7.69 (d, 2H, J = 8.7 Hz), 7.63 (s, 1H) , 7.50, (d, 1H, J = 7.2 Hz), 7.44 (dd, 1H, J - 8.7, 1.5 Hz), 7.16-7.22 (m, 2 H), 6.11 (s, 1H), 3.23 (br s, 2H), 2.55 (t, 2H, J = f > , - ss¿- ^ A i 222 The title compound was prepared in a 78% yield by means of the SEM deprotection of intermediate 55b in a manner analogous to procedure i used for example 41. H NMR (300 MHz, DMSO-de) d 13.95t < (s, ÍH), 13.04 (s, ÍH), 10.98 (s, ÍH), 9.54 (s, 1H), 9.30 (d, 2H, J * 1.8 Hz), 8.83 (dd, ÍH, J = 4.8, 1.8 Hz), 8.65 (s, ÍH), 8.59 (s, ÍH), 8.45-8.50 (m, 4H), 7.52-7.66 (m, 4H), 7.18 (br s, 2H). Analysis (C29Hi9N70 • 0.5H20) C, H, N. MS (ES) [m + H] / z calculated 482, found 482.
Example 56: N-. { 4- [3- (l-Benzo m? Dazol-2-yl) lg-? Ndazol-5-al] -isoqu? Nolm-8-? L} - acetamide [Dioxane = Dioxane] "* < = »223 (a) Intermediate 56a-N- (4-B? Fmf-soquinol? n-8-? l) -acetamide: 8-Am? no-4-bromo-? soqu? nolma (300 mg, 1.35 m ol ), DIEA (0.94 m, 5.38 mmol) and acetic anhydride (255 uL, 2.7 5 'mmol) were stirred in chloroform (20 ml) at reflux temperature for 16 hours. The solution was washed with and # brine, dried (Na 2 SO) and concentrated in vacuo. The residue was stirred in ethanol (6 ml) with HOAc (2 ml) at a temperature of 72 ° C for 20 hours. The ib * solution was either cooled or diluted and diluted with ethyl acetate. The organic compounds were washed with 1 N NaOH and brine, dried (MgSO 4), and concentrated in vacuo. Purification by # chromatography on silica gel (ethyl acetate) gave 232 mg (65%) of intermediate 56a ßn 15 as a white solid. XE NMR (300 MHz, CDCl 3) § 10.31 (s, 1H), 9.47 (s, ÍH), 8.77 (s, 1H), 7.90-7.97 (, 3H), 2.21 (s, 3H). Analysis (CnH9BrN20) C, H, N. (b) Intermediate substance 56b-27-. { 4-l3- (lH- Benzoi? N? Dazol-2-? L) -1- (2-tpmet? Ls? Lanil-efe? ExXymethyl) -lflA 0? Ndazol-5-? L] -? Soquinol? N -8-? L} Acetamide: The title compound was prepared in an 80% yield from the intermediate Z5a, and the intermediate 56a in a manner similar to the "procedure used for the intermediate 41a.lH 5 NMR (300 MHz, CDC13) d 10.39 (s, ÍH), 9.46 (s, ÍH), 8.76 (s, 2 ÍH), 8.53 (s, ÍH), 8.42 (s, 1H), 7.91 (d, ÍH, J = 7.2 Hz), 7.89 (d, ÍH, J = 7.2 Hz), 7.62-7.72 (m, 3H) , 7.48-7.57 (ra, 3H), 7.24-7.28 (m, ÍH), 5.83 (s, 2H), 3.65 (t, 2H, J - 8.1Hz), 2.35 (s, 3H), 0.95 (t, (c) Example 56-il) -lfi-indazol-5-yl] -isoquin The title compound was prepared in a "~ * 4. • - - - yield of 68% by means of the SEM deprotection of "intermediate 56b in a manner analogous to the procedure used for example 41. XH NMR (300 MHz, DMSO-de) d 13.84 (s, 1H) , 13.03 (s, ÍH), 10.34 (s, ÍH), 9.56 (s, 1H), 8.61 (s, ÍH), 8.55 (s, ÍH), 7.82-7.89 (m, 2H), 7.74 (t, 1H , "% ** J = 7.2 Hz), 7.59-7.66 (m, 3H), 7.51 (d, 1H, J = 7.2 f? Z), 7.13-7.21 (m, 2H), 2.25 (s, 3H). Analysis (C25H? 8N60 • 0.4H20) C, H, N. MS (ES) [m + H] / z calculated 419, found 419, Example 57: N-. { 4- [3- (l-Benzoimidazol-2-yl) lfl-i da2 l-5-% * ^ t jl] -isoquininolin-8-il} benzyl amine [Dioxane = Dioxane] t (a) Intermediate 57a-Benzyl- (4- 15-bromo-? soqu? nol? na-8-? l) -amine: 8-Am? no-4-bromo-? soqumol? na (220 mg, 0.99 mmol) and benzaldehyde (110 μl, 1.1 mmol) were stirred in ethanol (15 ml) / HOAc (0.2 ml) at the reflux temperature for 24 hours. The reaction was cooled to a temperature of 0 ° C, and sodium cyanoborohydride (622 mg, 9.9 mmol) was added in portions. After stirring for 1 hour, the reaction was diluted with H2G. and extract with ethyl acetate. The organic compounds were washed with brine, dried (Na 2 SO 4 and concentrated in vacuo.
The title compound was prepared in a < 72% yield by means of the SEM deprotection of id "" intermediate substance 57b analogously to the procedure used for example 41. H NMR (300 MHz, DMSO-de) $ 13.80 (s, ÍH), 13.01 (s, ÍH), 9.71 (s, 1H), 8.56 (s, ÍH), 8.42 (s, 1H ), 7.71-7.81 (m, 2H), 7.30-7.63 (m, 8H), 7.17- 7.25 (m, 3H), 6.90 (d, ÍH, J = 7.2 Hz), 6.52 (d, ÍH, J- 7.2 Hz), 4.57 (d, 2H, J = 5.4 Hz). Analysis (C3oH22N6 • 0.5 H2Q) C, H, N. MS (ES) [m + H] / z calculated 467, found 467. 10 Example 58: 3- [3- (l-Benzo m-dazol-2-yl) -H-ndazol * -5-l] -4-met l- [3, 3 '] b? P r? Dmil [Dioxane = Dioxane] 25 ? í $? (a) Intermediate 58a-5-Bromo-4-methyl- [3, 3 '] bipyridinyl: The title compound was prepared in a 54% yield from 3,5-d-bromo-4-met ? lp? r? dihd and 3-pipd? lonic acid in a manner analogous to the procedure * used for intermediate 53a. 1H NMR (300 MHz ,,, CDC13) d 8.67-8.71 (m, 2H), 8.59 (dd, ÍH, J * 2.4, 0.6 Hz), 8.33 (s, ÍH), 7.62-7.66 (m, ÍH), 7.39-7.44 (m, ÍH), 2.35 (m, 3H). Analysis (CnH9BrN2 • 0.1H2O) C, H, N. (b) Intermediate substance 58b-3- [3- (lff-Benzo? m? dazol-2-yl) -1- (2-trimethylamino? -ethoxymethyl) -Ifl-? ndazol-5-? l] -4-met? L- [3, 3 'Jbípiridinil: The title compound was prepared in a 37% yield from intermediate 25a and intermediate 58a in a manner analogous to the procedure used for intermediate 41a. H NMR (300 MHz, CDC13) d 10.24 (s, ÍH), 8.68-8.71 (m, 3H), 8.61 * (s, ÍH), 8.47 (s, ÍH), 7.85-7.88 (m, ÍH), 7.70 -7.78 (m, 2H), 7.42-7.53 (m, 3H), 7.26-7.33 (m, 2H), 5.83 (s, 2H), 3.64 (t, 2H, J - 8.1HZ), 2.19 (s, 3H) ), 0.95 (t, 2H, J - 8.1Hz), -0.05 '(s, 9H). 4 - (c) Example 58 -3- [3- (1-Benzoimidazol-2-yl) -l, -mdazol-5-yl] -4-methyl- [3, 3 '] bipyridyl: The title compound was prepared in a yield of 74% by means of the deprotection SEM of the intermediate substance 58b analogously to the procedure used for example 41. lE NMR (300 MHz, DMSO-d «) d 13.79 ( s, ÍH), 13.02 (s, ÍH), 8.72 (d, 1H, J -.1.5 Hz), §, 65 (dd, ÍH, J - 4.8, 1.5 Hz), 8.54 (s, ÍH), 8.52 ( s, 1H), 8.48 (s, ÍH), 7.96-8.00 (m, ÍH), 7.78 (d, ÍH, J - 8.7 Hz), 7.6S (d, ÍH, J = 7.5 Hz), 7.49- 7.57 (m, 3H), 7.17-7.23 (m, 2H), 2.16 (s, 3H). Analysis (C25H? 8N6 • 0.6H20) C, H, N. MS (ES) [m + H] / z calculated 403, found 403.
Example 59: (E) -3-. { 5- [3- (LFI-Benzoimidazo > l- til) -lfí-indaiol-5-yl] -4-methyl-pyridin-3-yl} -prop-2-in-l-ol ? je plo 60: (E) -3-. { 5- [3- (1-Benzyl] imidazol-2-yl) -lff-inda-1 -5-iX] -4-methylpyridin-3-yl} -propan-l-ol [Pipepdme = Piperidma Pypdme = Pyridine Reflux = Reflux Dioxane = Dioxane] (a) Intermediate 59a-5-Bromo-4- met? l-pyr? dma-3-carbaldehyde: 3,5-D? bromo-4-met? lp? pd? na (3.8 g, 15.1 mmol) s? stirred in dry THF (150 ml) at a temperature of -100 ° C (N2 / ether) in argon. N-Butyllithium (2.5 M) was added dropwise Hz), 4.29 (q, 2H, J «7.2 Hz), 2.50 (s, 3H), 1.35 (t, 3H, p> = 7.2 Hz). Analysis (CnH12BrN02) C, H, N. (c) Intermediate substance 59c - (E) -3-. { 5- [3- (1-Benzoimidazol-2-yl) -1- (2-trimethylsilanyl-ethoxymethyl) -lfl-indazol-5-yl] -4-methyl-pyridin-3-yl} ethyl ester of acrylic acid: The title compound was prepared in an 83% yield from intermediate 25a and intermediate 59b in a manner similar to the procedure used for intermediate 41a. iH, NMR (300 MHz, CDC13) d 10.00 (s, ÍH), 8.74 (s, ÍH), 8.65 (s, ÍH), 8.55 (s, ÍH), 7.99 (d, ÍH, J = 15.9 Hz), 7.83- 7.87 (m, ÍH), 7.71 (d, 1H, J = 8.7 Hz), 7.50-7.53 (m, ÍH), 7.43 (dd, ÍH, J - 8.7, 1.5 Hz), 7.27-7.32 (m, 2H) , 6.50 (d, ÍH, J «15.9 Hz), 5.84 (s, 2H), 4.31 (q, 2H, J = 7.2 Hz), 3.65 (t, 2H, J = 8.1Hz), 2.36 (s, 3H), 1.37 (t, 3H, J - 7.2 Hz), 0.95 (t, 2H, J = 8.1Hz), -0.04 (s, 9H). (d) Intermediate substance 59d- (E) -3-. { 5- [3- (IHr Benzoimidazol-2-yl) -1- (2-trimethylsilanyl-ethoxymethyl) - x J.fí-? Ndazol-5-yl] -4-met? Lp? Rid? N-3-il } -prop-2-en-l-ol: A solution of intermediate 59c (402 mg, 0.73 mmol) in ether (10 ml) was added dropwise to a stirred suspension of LAH (180 mg, 4.74 mmol) in ether (10 ml) at a temperature of 0 ° C. He let the -4 > ísS # fef «'' EfCWÍ reaction for 3 hours while warming it to room temperature. The reaction was tempered with water and? it was extracted with ethyl acetate. The organic compounds were washed with brine, dried over Na 2 SO 4, and concentrated in vacuo. Purification by chromatography on silica gel (50% to 100% ethyl acetate / hexanes) provided 72 mg (19%) of intermediate 59d as a white foam (followed by 186 mg (50%) ) of intermediate '59e) XE NMR (300 MHz, CDC13) d 9.99 (s, ÍH), 8.64 (s, ÍH), 8.63 (S, 1H), 8.45 (s, ÍH), 7.83-7.87 (m , ÍH) - 7.69 (d, ÍH, J = 8.7 Hz), 7.50-7.53 (m, ÍH), 7.43 (dd, ÍH, J = 8.7, 1.5 Hz), 7.26-7.32 (m, 2H), 6.86 ( d, ÍH, J - 15.9 Hz), 6.33-6.41 (m, 1H), 5.84 (s, 2H), 4.42 (br s, 2H), 3.65 (t, 2H, J = 8.1Hz), 2.28 (s, 3H), 1.73 (br s, 1H), 0.95 (t, 2H, J = 8.1Hz), -0.04 (s, 9H). (e) Intermediate substance 59e - (E) -3-. { 5- [3- (1-Benzoyl? Dazol-2-? L) -1- (2-trimethylaryl-ethoxymethyl) -lfl-mdazol-5-? L] -4-methyl-pyridin-3? l) - propan-1-ol: See the procedure used for the intermediate substance 59d above. XE NMR (300 MHz, CDCl 3) d 10.17 (s, ÍH), 8.63 (s, ÍH), 8.40 (s, 2H), 7.83-7.87 (m, 1H), 7. ^ 68 (d, ÍH, J = 8.7 Hz), 7.49-7.52 (m, ÍH), 7.43 (dd, IB, J - 8.7, 1.5 Hz), 7.26-7.31 (m, 2H), 5.83 (s, 2H), 3.77 (t, 2H, -"' 2. 3. 4 J = 6.3'Hz), 3.65 (t, 2H, J = 8.1Hz), 2.82 (t, 2H, J - 7.5 Hz), 2.24 (s, 3H), 1.88-1.95 (m, 2H), 1.74 (br s, ÍH), 0.94 (t, 2H, J - 8.1Hz), -0.05 (s, 9H). (f) Example 59 - (E) -3-. { 5- [3- (1 * -B < Snzoimidazol-2-yl) -lH-indazol-5-yl] -4-methyl-pyridin-3-yl} -prop-2-en-l-ol: The title compound was prepared in a 50% yield by means of the SEM deprotection of intermediate 59d in a manner analogous to the procedure used for example 41. aH NMR (300 MHz , DMSO-de) d 13.78 (s, ÍH), 13.02 (s, ÍH), 8.63 (s, ÍH), 8.43 (s, ÍH), 8.35 (s, ÍH), 7.75 (d, ÍH, J = 8.7 Hz), 7.63 (br. S, 1H), 7.55 (br s, ÍH), 7.46 (dd, ÍH, J - 8.7, 1.5 Hz), 7.20 (br s, 2H), 6.83 (d, ÍH, J - 15.9 Hz), 6.37-6.46 (m, 1H), 4.99 (t, ÍH, J = 5.4 Hz), 4.19 (s, 2H), 2.23 (s, 3H). Analysis (C23H19N5O «0.6 H20) C, H, N. MS (ES) [m + H] / z calculated 382, found 382. (g) Example 60 - (E) -3-. { 5- [3- (L-, Benzoimidazol-2-yl) -lfl-mdazol-5-yl] -4-methyl-pyridm-3-yl} -propan-1-ol: The title compound was prepared in a 62% yield by means of the SEM deprotection of intermediate 59e in a manner analogous to the procedure used for example 41. XH NMR (300 MHz, DMSO-de ) d 13.77 (s, 1H), 13.02 (s, ÍH), 8.41 (S, ÍH), 8.37 (s,? H), 8. 30 (s, ÍH), 7.73 (d, 1H, J * 8.7 Hz), 7.67 (br s, ÍH), 7.52 (br s, ÍH), 7.44 (dd, ÍH, J = 8.7, 1.5 Hz), 7.19 (br s, -2H), 4.57 (t, ÍH, J - 5.1Hz), 3.49 (q, 2H, J - 6.0 Hz),, 2.74"* y * (t, 2H, J - 7.8 Hz), 2.21 (s, 3H), 1.69-1.79 (m, 2H). ^ í Analysis (C23H21N5O • 0.5H20) C, H, N. MS (ES) [? FH-H] / z * calculated 384, found 384.
Example 61: 5- [3- (1-Benzo? M dazol-2-? L) - > lí-ijr > dazol-5-i l * 4-r et? l- [3,4 '] b? p? r? d? n? (a) Intermediate 61a-3, 5-Dβ-brom-4-et l-pyridine: The title compound was prepared in a yield of 74% by the substitution of iodoethane with iodomethane in the process for the preparation of 3,5- ít £ ** It was concentrated in vacuo. The on silica gel (75% a gave 232 mg (60%) of the intermediate substance 61c ^ form of a clear colored oil. 1H NMR (300 MHz, CDC13) d 10.18 (s, ÍH), 8.71-8.75 (m, 3H), 8.57 (s, ÍH), 8.41 (s, ÍH), 7.84-7.87 (m, ÍH), 7.71 ( d, ÍH, J - 8. Hz), 7.48- 7.53 (m, 2H), 7.36 (dd, 2H, J = 4.5, 1.5 Hz), 7.26-7.32 (, 2H), 5.84 (s, 2H), 3.65 (t, 2H, J- 8.1Hz), 2.64 (q, 2H, J -7.5 Hz), 0.94 (t, 2H, J «8.1Hz), 0.77 (t, 3H, J = 7.5 Hz), - 0.04 ( s, 9H). (d) Example 61-5- [3- (1-Benzoimidazol-2-yl) -lfi-anda2ol-5-? l] -4-et? l [3, 4 '] b? p? r? din? 1: The title compound was prepared in a 61% yield by means of the SEM deprotection of intermediate 61c, analogous to the procedure used for example 41. 1 H NMR (300 MHz, DMSO-de) d 13.79 (s, 1H), 13.02 (s, ÍH), 8.71 (dd, 2H, J = 4.5, 1.5 Hz) , 8.51 (s, 2H), 8.40 (s, ÍH), 7.78 (d, ÍH, J * 8.7 Hz), 7.68 (d, ÍH, J = 7.5 Hz), 7.49-7.56 (m, 4H), 7.14- 7.25 (m, 2H), 2.59 (q, 2H, J - 7.5 Hz), 0.69 (t, 3H, J = 7.5 Hz). TAnállsiS (C26H20N6 • 0.3H20) C, H, N. MS (ES) [m + H] a calculated 417, found 417.
Example 62: 3- [3- (1-Benzooim? Da2ol-2-? L) -lfi-imtazol-5-yl] -4- - *. .., j - * ft 6 methyl- [2, 3 '] bipyridine? [Sulfur = Sulfur Dioxane = Dioxan Anisol = Anisol] (a) Intermediate 62a-3- (l-Benzoimidazol-2? l) -5-iodo-l- (4-m tox? -benzyl) -lfl? nda2?: The title compound was prepared in a yield of 59% from intermediate 19d and phenylenediamine in a manner similar to the procedure used for intermediate 7c '. ? E NMR (300 MHz, DMSO-ds) d 13.06 (s, ÍH), 8.91 (s, ÍH), 7.70-7.78 (m, 3H), 7.51 (dd, 239 ÍH, J - 6.3, 2.1Hz), 7.19-7.28 (m, 4H), 6.88, (dd, 2H, J = 6.6, 2.1Hz), 5.72 (s, 2H), 3.69 (s, 3H). Analysis (C22H? 7IN40) C, H, N. (b) Intermediate 62b-3 * - (lff-Benzoimidazol-2-yl) -l- (4-methoxy-benzyl) -5- (4,5,5-tetramethyl [1, 3, 2] dioxaborolan-2 -il) -lfí-indazol: The title compound was prepared in a 73% yield from the substance in a manner analogous to the preparation of 'intermediate 19e. XH NMR (300 MHz, DMSO-de) d 13.03 (s, 1H), 8.93 (d, ÍH, J - 4.2 Hz), 7.78-7.84 (m, 2H), 7.73 (dd, ÍH, * J = 8.7, 0.9 Hz), 7.51 (d, ÍH, J = 7.2 Hz), 7.20-7.27 (m, 4H), 6.87 (d, 2H, J «8.7 Hz), 5.74 (s, 2H), 3.68 (s, 3H), 1.34 (s, 12H). Analysis (C28H29BN403) C, H, N. $ (c) Intermediate 62c-5 '-Bromo- * f-methyl- [2, 3' J bipyridinyl: p. 3, 5-Dibromo-4-met? L-pyridine (2.0 g, 7.8 mmol) and 2-tributyltanamlpyridine (2.4 g, 6.5 mmol) were shaken nn? Oxano (20 ml) in a purged beaker with argon. 0 Tetrakis (triphenylphosphine) -palladium (0) (600 mg, * 0.5 mmol) was added and the reaction was stirred at a temperature of 100 ° C for 80 hours. The solution was concentrated in vacuo and purified by chromatography on silica gel (30% to 50% ethyl acetate / hexanes, two purifications) to provide 788 mg (49%) of intermediate 62c as a white solid. ? E NMR (300 MHz, CDC13) 8.72-8.75 (m, ÍH), 8.70 (s, ÍH), 8.46 (s, 1H), 7.78-7.84 (m, ÍH), 7.31-7.42 (m, 2H) , 2.42 (s, 3H). Analysis (CuH9BrN2) C, H, N. f (d) Intermediate 62d-3- [3- (lfl * -Benzoimidazol-2-yl) -1- (4-methoxy-benzyl) -lfi-indazol-5- il] -4- ethyl- [2, 3 '] bipyridini1: The title compound was prepared in a 76% yield from intermediate 62b, and intermediate 62c similarly to the "procedure employed for intermediate 61c. XH -NMR (300 MHz, DMSO-d6) d 13.08 (s, ÍH), 8.73 (d, ÍH, J = 4.2 Hz), 8.52-8.57 (m, 3H), 7.92-7.98 (m, 2H), 7.68 -7.72 (m, 2H), 7.58 (dd, ÍH, J - 8.7, 1.5 Hz), 7.51 (d, ÍH, J = 7.2 Hz), 7.43-7.47 (m, ÍH), 7.36 (d, 2H, J = 8.7 Hz), 7.17-7.23 (m, 2H), 6.91 (d, 2H, J = 8.7 Hz), 5.79 (s, 2H), 3.70 (á, 3H), 2.21 (s, 3H). Analysis (C33H26N6O) C, H, N. (e) Example 62 -3- [3- (1-Benzoimidazol-2-yl) -lfl-ipdazdl-5-yl] -4-met? l- [2, 3 '] bipyridinyl: Intermediate 62d (400 mg, 9.77 mmol) was stirred in a solution of concentrated H2SO4 (1 ml) and anisole (1 * ml) in TFA (8 ml) for 48 hours. The solution was concentrated to a volume of ~ 3 ml in vacuo and then quenched with NaHCO3"pi *. * »« ~ I i 2 * • h * Saturated and extracted with ethyl acetate / THF in a ratio of 4: 1. The organic compounds were washed with brine, dried (MgSO 4) and concentrated in vacuo. Purification by chromatography on silica gel 0.% " NH40H / 6% to 10% MeOH / ethyl acetate) yielded 102 mg (33%) of the substance of Example 62 in the form of a white solid. XE NMR (300 MHz, DMSO-de) d 13.78 (s, ÍH), 13. 02 (s, 1H), 8.74 (d, ÍH, J = 4.2 Hz), 8.57 (s, 1H), 8.53 (s, ÍH), 7.51 (s, ÍH), 7.93-7.99 (m, ÍH), 7.77 (d, ÍH, J = 8.7 Hz), 7.70 (d, 2H, J = 7.8 Hz), 7.43-7.56 ( m, 3H), 7.20 (br s, 2H), 2.23 (s, 3H). Analysis (C25H? 8N6 • 0.5 H20) C, H, N. MS (ES) [m + H] / z calculated 403, found 403.
Example 63: l-. { 5- [3- (1-Benzoimidazol-2-yl) -fí-indagol-5-yl 3 ^ 3, 4-dihydro-2-yl [1,7] naphyridin-1-yl} -etanone •%. 243 (b) Intermediate 63b-1- (5 ~ B-OHOH ~ 3,4-dihydro-2-y [1, 7] naphthyrid? n-1-yl) ethanone: Intermediate 63a (212 mg, 1.0 m ol) ), DIEA (1.4 m, 8.0 mmol) and acetic anhydride (4.0 mraol) were stirred in dry chloroform (10 ml) at a temperature of ', 68 ° C for 40 hours. The solution was washed with saturated NaHCOs and brine, dried (Na2SO4) and concentrated in vacuo. Purification by chromatography on silica gel (70% ethyl acetate / hexanes) provided 242 mg (95%) of the intermediate 63b as a white solid. XH NMR (300 MHz, CDC13) d 8.61 (br s, ÍH), 8.46 (s, ÍH), 3.79 (q, 2H, J = 6.0 Hz), 2.83 (t, 2H, J = 6.9 Hz.}., 2.93 (s, 3H), 2.01-2.08 (m, 2H) Analysis (C1QHnBrN20) C, H, N. (c) Intermediate substance 63c-1-. { 5- [3- (1-Benzoimidazol-2-yl) -1- (2-trimethylsilanylethyloxymethyl) -lfl-indazol-5-yl] -3,4-dihydro-2-yl] [1,7] naphthyridin-1-yl } - ethanone: - s The title compound was prepared in an 83% yield from intermediate 25a and intermediate 63c in a manner similar to the procedure used for intermediate 61c aH NMR (300 MHz, CDC13) d 10.06 (s, ÍH), 8.68 (s, ÍH), 8.55 (br s, ÍH), 8.42 (s, ÍH), 7.84-7.87 (m, ÍH), 7.71 (d, ÍH, J - 8.7 Hz), 7.50-7.53 (m, ÍH), 7.46 (dd, ÍH, J = 8.7, 1.5 Hz), 7. 2? -7.32 (m, 2H), 5.84 (s, 2H), 3.84 (t, 2H, J - 6.6 Hz), 3.64 (t, 2H, J = 8.1Hz), 2.70 (t, 2H, J - 6.6 -Hz), 2.36 (S, 3H), 1.87-1.93 (m, 2H), 0.94 (t, 2H, J = 8.1Hz), -0.04 (S, 9H). Analysis (C3oH34Ne02Si «0.5 H20) C, H, N. (d) Example 63-1- (5- [3- (1-Benzimidazol-2-yl) -lfl-indazol-5-yl] -3,4-dihydro-2-yl [1, 7] naphthyridin-1 - il.} - ethanone: The title compound was prepared in a 65% yield by means of the SEM deprotection of the intermediate 63c in a manner analogous to the procedure used for example 41. 1 H NMR (300 MHz, DMSO -de) d 13.77 (s, ÍH), 13.02 (s, ÍH), 8.78 (s, ÍH), 8.46 (br s, ÍH), 8.27 (s, ÍH), 7.75 (d, 1H, J = 8.7 Hz ), 7.67 (br. S, ÍH), '' 7.49 (dd, 2H, J = 5.7, 1.5 Hz), 7.20 (br s, 2H), 3.74 (t, 2H, J = 6.3 Hz), 2.63 (t , 2H, J - 6.3 Hz), 2.27 (s, 3H), * 1. 79-1.85 (m, 2H). Analysis (C24H20NeO) C, H, N. MS (ES) [m + H] / z calculated 409, found 409.
Ejepplo 64: 5- [3- (lH-Benzoimidazol-2-yl) -lH-indazol ^ -5 * - il] -4-methyl-nicotinamide Chromatography on silica (ethyl 5: 1 to 10: 1 hexanes-acetate and then with 100% ethyl acetate) yielded 236 mg (9%) of the 64a amide. Rf = 0.09 (50% ethyl acetate in hexanes); XH NMR (300 MHz, CDC13) d 8.70 (bs, ÍH), 8.54 (bs, 1H), 5.98 (bs, ÍH), 5.93 (bs, ÍH). (LCMS: M + 215). (b) Intermediate substance 64b-5- [3- < L-Benzoim? dazol-2-yl) -1- (2-trimethylsilylethylethoxymethyl) -lfl-indazol-5-yl] -4-methyl-nicotinamide: The title compound was prepared in a 75% yield from of intermediate 25a and intermediate 64a in a manner similar to the procedure used for intermediate 61c. 1H NMR (300 MHz, CDC13) d 10.18 (s, ÍH), 8.66 (s, ÍH), 8.64 (s, 1H), 8.59 (s, ÍH), 7.82-7.86 (m, ÍH), 7.70 ( d, 1H, J = 8.4 Hz), 7.48-7.52 (m, ÍH), 7.42 (dd, ÍH, J «8.4, 1.5 Hz), 7.26-7.31 (m, 2H), 6.13 (br s, ÍH), 5.91 (br s, ÍH), 5.83 (s, 2H), 3. * 64 (t, 2H, J «8.1Hz), 2.40 (s, 3H), 0.94 (t, 2H, J - 8.1Hz), - 0.04 (s, 9H). (c) Example 64-5- [3- (1-Benzo [mu] -dazol-2-yl) -lfl-indazol-5-yl] -4-methylnicotinamide: The title compound was prepared in a yield of 77% by means of the SEM deprotection of the intermediate substance 64b analogously to the procedure used for example 41. XH NMR (300 MHz, DMSO-de) d 13. 80 (s, 1H), 13.03 (s, ÍH), 8.54 (s, 1H), ß.50 (s, 1H), "f '8.45 (s, 1H), 8.05 (s, ÍH), 7.76 (d) , 1H, J * 8.7 Hz), 7.68 (br, s, 2H), 7.44-7.52 (m, 2H), 7.16-7.22 (m, 2H), 2.29 s,. 'R 3H) Analysis (C2iHi6N60 • 0.55 H20) C, H, N. MS (ES) [m + Ji] / z; calculated 369, found 369.
Example 65: 3- (1-Benzoimidazol-2-yl) '-5- [5- (1-methyl-4-yl) -4-methyl-1-pyridin-3-yl) -lfi-indazole 1? 65a 65b (a) Intermediate 65a-3-Bromo-4-methyl-5-lfl-imidazole-4? -pyr? dma: To a stirred suspension of tosylmethyl isocyanide (1.02 g, 5.25 mmol) and 3- bromo-4-met l-5-formyl pyridine (1.0 g, 5 mmol) in 5 ral of dry ethanol was added finely powdered NaCN (25 mg, 0.5 mmol) to a *. ,, x 248 temperature of 25 ° C. After 30 minutes, the reaction was concentrated in an oil. The resulting oil was added to a saturated solution of ammonia in dry methanol in a sealed tube and heated to 100 ° C for 24 - '/ hours. Cooling and concentration followed by chromatography on silica (10: 1 ethyl acetate-hexanps) provided 167 mg (14%) of intermediate 65a as a white solid. XE NMR (300 MHz, DMSO-d6) 12.45 (bs, ÍH), 8.71 (s, 1H), 8.55 (s, ÍH), 7.83 (S / ÍH), '"7.52 (s, 1H), 2.56 (s) , 3H). (b) Intermediate 65b-3-Bromo-4-methyl-5- [1- (2-trimethylsilanyl-ethoxymethyl) -lfi-imidazol-4-yl] -pyridine: Intermediate 65a was protected with SEM in a yield 45% in a manner analogous to the procedure used for intermediate 49a. 1H NMR (300 MHz, CDC13) d 8.67 (s, ÍH), 8.60 (s, 1H), 7.70 (s, ÍH), 7.21 (d, ÍH, J - 1.2 Hz), 5.33 (s, 2H), 3.56 (t, 2H, J = 8.1Hz), 2.58 (s, 3H), 0.94 (t, 2H, J = 8.1Hz), 0.00 (s, 9H). Analysis (C? 5H22BrN3OSÍ) C, H, N. (c) Intermediate 65s-3- (1-J2-imidazol-2-yl) -5-. { 5- [1- (2-Trimethyl-silyl-α-toxymethyl) -lfi-imidazol-4-yl] -4-methyl-pyridin-3-yl} -1- (2-trimethylsilanyl-ethoxymethyl) -lfi-indazole: ,? ^ -,. 249 f The title compound was prepared in an 83% yield from intermediate 2 & amp; and intermediate 65b in a manner similar to the procedure employed for intermediate 61c. 1H NMR (300 MHz, CDC13) d 10.09 (s, 1H), 8.85 (s, ÍH), 7.70 (S, ÍH), 8.50 (s, ÍH), 7.84-7.88 (m, ÍH), 7.68-7.74 ( m, 2H), 7.47-7.52 (m, 2H), 7.26-7.31 (m, 3H), 5.84 (s, 2H), 5.36 ($ | 2H), 3.55-3.68 (m, 4H), 2.41 (s, 3H), 0.92-0.98 (m, 4H), -0.01 (s, 9H), -0.05 (s, 9H). (d) Example 65 -3- (1-Benzoimidazol-2-yl) -5- [5- (1-yl-imidazol-4-yl) -4-met? lp? ridin-3? l] -lfl-indazole : > The title compound was prepared in a 43% yield by means of the SEM deprotection of the intermediate 65c in a manner analogous to the procedure used for example 41. XH NMR (300 MHz, DMS0-d6) d 13.78 (s, ), 13.02 (s, ÍH), 12.38 (s, ÍH), 8.83 (s, 1H), 8.47 (s, ÍH), 8.35 (s, ÍH), 7.83 (d, ÍH, J - 0.9 Hz), 7.76 (d, ÍH, J = 8.4 Hz), 8.69 (d, 1H, J = 7.5 Hz), 7.47-7.51 (ra, 3H), 7.16-7.22 (m, 2H), 2.37 (s, 3H). Analysis (C23H17N7 • 2 «5 * 'H20) C, H, N. MS (ES) [m + H] / z calculated 392, found 392.
Example 66: 4- [3- (4, 5, 6, 7-Tetrah? Dro-lfíj »bew &oimidago3.« - 2- jl) -lfí-mdazol-4 -? L] isoquinoline (b) Intermediate 66b-4- [3- (4, 5,6,7-, 1-tetrahydro-1-benzyl-2-yl-2-yl-2-yl) -1- (2-tnmethylsilane-ethoxymethyl) -lfi -mda2? l-5-yl] -isoqumolma: The title compound was prepared in 75% yield from intermediate 6 a and. and acid? soqumolm-4-boron? co (EP 976747) similarly to the procedure used for intermediate 7d1, XH NMR (300 MHz, CDC13) d 9.57 (br s, ÍH), 9.28 (s, ÍH), 8.67 (s, ÍH), 8.58 (s, ÍH), 8.04-8.08 (m, 1H), 7.85-7.89 (s, 1H), 7.57-7.70 (m, 4H), 5.80 (s, 2H), 3.63 ( t, 2H, J - 8.1Hz), 2.7 (br s, 4 H), 1.86 (br s, 4 H), 0.96 (t, 2 H, J = 8.1 Hz) * - '0.03 (s, 9 H). (c) Example 66-4- [3- (, 5,6,7-? ehydro-l-benzyl-2-yl-2-? l) -lfl-nda2? l-5-yl] -isoquinoline: The title compound was prepared in 64% yield by means of the SEM deprotection of intermediate 66b in a manner analogous to the procedure used for Example 41. X H NMR (300 MHz, DMSO-de) d 13.31 (s, ), 12.25 (s, ÍH), 9.36 (s, ÍH), 8.49 (s, 1H), 8.43 (s, ÍH), 8.24 (d, 1H, J = 7.8 Hz), 7.69-7.85 (m, 4H) , 7.53 (dd, 1H, J - 8.7, 1.8 Hz), 2.50 (br s, 4H), 1.73 (br s, 4H). Analysis (C23H19N5 • 0.2H20) C, H, N. MS (ES) [m + H] / z Calculated 366, found 366.
F" Example 67: 4- [3- (4-Met? L-5-f in l-l-ymidazole-2-? L) - > lfí- '? ndazol-5-3. soqumol na [Benzene = Benzene Reflux = Anisol Reflux = Anisole] (a) Intermediate substance 67a-5-Iodo-l- (4-methoxy-benzyl) -3- (4-met? l-5-phen? l-lf? -m? dazol-2-? l) -lfi mdazole: The title compound was prepared from intermediate 19d and 1-phen? ll, 2-propanedione in a manner similar to the procedure used for intermediate 66a. XH NMR (300 MHz, CDC13) d 9.87 (br s, 0.5 H), "9.71 (br s, 0.5 H), 8.98 (br s, 0.5 H), 8.92 (br s, 0.5 H), 0 7.84 (d) , 1H, J = 7.2 Hz), 7.61 (dd, ÍH, J = 8.7, 1.5 Hz), 7.44-7.53 £ m, 3H), 7.31 (d, ÍH, J = 7.5 Hz), 7.11 (app d, 3H , J = 8.7 Hz), 6.81 (dd, 2H, J - 6.6, 1.8 Hz), 5.49 (s, 2H), 3.77 (s, 3H), 2.55 (s, 3H) Analysis (C25H2IN4?) C, H , N. 5 (b) Intermediate substance 67b-4- [1- (4-MefcF? i-benzyl) * S- (4-methyl-5-phenyl-1-yl-imidazol-2-yl) -% N-indazol-5-yl ] -isoquinoline: The title compound was prepared in a 75% yield from intermediate 67a and isoquinolm-4-boron acid co (EP 976747) analogously to the procedure used for intermediate 7d '. XH NMR (300 MHz, CDC13) d 9.97 (br s, 0.5 H), 9.85 (br S, 0.5 H), 9.28 (s, HI), 8.76 (br s, 0.5 H), 8.70 (br s, 0.5 H) ), 8.59 (s, 1H), 8.03-8.10 (m, ÍH), 7.90 (br s, 1H), 7.75 (br s, ÍH), 7.37-7.68 (m, 8H), 7.20-7.26 (m, 2H) ), 6.86 (dd, 2H, J = 6.6, 1.5 Hz), 5.60 (s, 2H), 3.78 (s, 3H), 2.54 (br s, 1.5H), 2.49 (br s, 1.5H). (c) Example 67-4- [3- (4-Methyl-5-phenyl-1-yl-imidazol-2-yl) -lfl-indazol-5-yl] -isoquinoline: The title compound was prepared in a yield of 15% by means of the PMB protection of intermediate 67b in a manner analogous to the procedure used for example 62. XH NMR (300 MHz, DMSO-de) d 12.67 (s, ÍH), 13.02 (s, ÍH) , 9.38 (s, ÍH), 8.57 (s, ÍH), 8.53 (s, 1H), 8.25 (d, ÍH, J = 7.5 Hz), 7.66-7.91 (m, 6H), 7.57 (dd, ÍH, J - 8.7, 1.5 Hz), 7.30-7.33 (m, 2H), 7.15-7.18 (m, ÍH), 2.50 (s, 3H). Analysis (C 26 H 19 N • 0.5 H 20) C, H, N. S (ES) [m + H] / z calculated 402, found 402. faith • * 254 E em lo- 68: D methyl-. { 2- [5 * i £ ¡«g¡ ^ t -E., 4 '] bip ridinil-5- 1, i il) -lfí-indazol-3-yl] -lfí- "ber? zoimidazol-4-A ^ - 'itethyl] -amjftjt (a) Intermediate 68a- (2- [5-Iodo-l- (4-ethoxy-benzyl) -lfl-ndazol-3-yl] -lfi-benzoim-dazol-4-ylmethyl} -methanol: The title compound was prepared in a 40% yield from intermediate 19d and intermediate 23b in a manner similar to the procedure used for intermediate 7c.
NMR (300 MHz, DMSO-de) d 13 8.91 (s, ÍH), 7.64-7.77 (m, 1.5 Hz), 7.18-7.27 (m, 4H), (s, ÍH), 5.71 (s, ÍH), 5.14 5. 7 Hz), 4.86 (d, ÍH, J = 5.7 Hz), 3.69 (s, 3H) 1. (b) Intermediate substance 68b -. { 2- [5-Iodo * l- * 4-toxy-benzyl] -lfl-indazol-3-yl] -lfi-benzoimidazol-4-yl-ethyl} -dimethylamine: • Intermediate 68a (2.5 g, 4.9 ramol) and -DIEA (1.38 m, 10 ramol) were stirred in THF (90 ml) at a temperature of 0 ° C. Methanesulfonyl chloride (0.76 g) was added. m, 9.8 mmol), and the reaction was stirred for 2.5 hours at a temperature of 0 ° C. Dimethylamine was bubbled through the solution for 1 minute and the reaction was stirred for 2 hours while warming room temperature. The solution was warmed with H20 and extracted with ethyl acetate. The organic compounds were washed with saturated NaHC 3 3 and brine, dried (Na 2 SO 4) and concentrated in vacuo. Purification by chromatography on * and on silica gel (0.2% NH40H / 3% MeOH / ethylpV '*' acetate gave 2.56 g (97%) of intermediate 68b "as a white foam. XH NMR (300 MHz, CDCl 3) S 9.07 (d, ÍH, J = 0.9 Hz), 7.80 (d, ÍH, J - 7.8 Hz), 7.63 (dd, 1H, J = 8.7, 1.5 Hz), 7.07-7.25 (m, 5H), 6.85 (dd, H, t.
J = 6.6, 1.8 Hz), 5.61 (s, 2H), 3.77 (app s, 5H), 2.33 (s, 6H). (c) Intermediate substance 68c -. { 2- [l- (4- *. * Methoxy-benzyl) -5- (4, 4, 5,5-tetramethyl- [1, 3, 2] dioxaborolan-2? L) -lfl-ndazol-3? L] -lfl-benzoimidazole-i, 4-1-methyl-dimethyl- amine: The title compound was prepared in a 62% yield from the intermediate substance * ß8b similar to the method used for the preparation of the intermediate substance 19e. XH NMR (300 MHz, CDC13) d 9.11 (s, », * ÍH), 7.82 (dd, 2H, J = 8.4, 0.9 Hz), 7.35 (dd, 1H, J» 8.4, 0.9 Hz), 7.16-7.21 (m, 4H), 6.85 (dd, 2H, J = 6.9, 1.8 H), 5.64 (s, 2H), 3.80 (br s, 2H), 3.76 (s, 3H), 2.35 (s, 6H) , 1.37 (s, 12H). 15 (d) Intermediate substance 68d - Dimet? L-. { 2- [5- > (4-methyl- [3,4 '] bip? Rid? Nil-5-yl) -1- (4-methoxy-benzyl) -l-yl-indazol-3-yl] -lfi-benzoimidazol-4-yl-met ? l} -amine: The title compound was prepared in an 85% yield from intermediate 68c 0 and intermediate 53a in a manner similar to the procedure used for intermediate 61c. 1H «MR (300 MHz, CDC13) d 8.69-8.75 (m, 4H), 8.57 (s, ÍH), 8.43 (s, ÍH), 7.75 (d, ÍH, J = 8.1Hz), 7.46 (d, ÍH) , J = 8.7 Hz), r 7.34-7.40 (m, 3H), 7.25-7.28 (m, ÍH), 7.19 (t, ÍH, J = 7.6 X¿2'k (a) Intermediate 69a- [3- (5-Bromo-4-met? lp? r? dm-3-? l) -phenyl] -methanol: The title compound was prepared in a 79% rebirth from of 3, 5-d? bromo-4-met? l? r? d? na and 3 (hydroxymethyl) -feml-boronic acid in a manner similar to the procedure used for intermediate 53a. H NMR (300 MHz, CDC13) d 8.62 (s, ÍH), 8.22 (s, 1H), 7.42-7.46 (m, 2H), 7.29 (s, ÍH), 7.16-7.20 (m, 1H), 4.76 ( d, 2H, * J = 5.7 Hz), 2.48 (t, ÍH, J = 5.7 Hz), 2.32 (s, 3H). Analysis (C ^ 3H12BrNO «0.2 H20) C, H, N. (b) Intermediate 69b - (3- { 5- [3- (l-Benzo? m? dazol-2-? l) -1- (4-methox? -benzyl) -lfl-mdazol-5- ll] -4-met? lp? r? dm-3-? l.}. - phenyl) -methanol: The title compound was prepared in an 83% yield from intermediate substance 62b and intermediate 69a in a similar manner to the procedure used for intermediate 61c. 1H NMR (300 MHz, CDC13) d 10.39 (s, ÍH), 8.69 (s, ÍH), 8.45 (s, ÍH), 8.36 (s, ÍH), 7.83-7.87 (m, ÍH), 7.36-7.49 ( m, 7H), 7.22-7.31 (m, 4H), 6.84 (d, 2H, J = 8.7 Hz), 5.60 (s, 2H), 4.79 (s, 2H), 3.76 (s, 3H), 2.51 (br s, ÍH), 2.11 (s, 3H). (c) Example 69- (3-. {5- [3- (1-Ben-2'-imidazol-2-yl) -lfl-indazol-5-yl] -4-methyl-pyridin-3-yl} .alpha.-methanol: The title compound was prepared in a 41% yield by means of PMB deprotection of intermediate substance 69b analogously to the procedure used for Example 62. 1H NMR (300 MHz, DMSO-d6 ) d 13.79 (s, ÍH), 13.02 (s, ÍH), 849 (d, 2H, J - 6.3 Hz), 8.41 (s, ÍH), 7.76 (d, ÍH, J = 8.7 Hz), 7.34-7.69 (m, 7H), 7.19-7.22 (m, 2H), 5.25 (br s, 1H), 4.58 (s, 2H), 2.15 (s, 3H). Analysis (C27H21N502 • 1 .2H20) C, H, N. MS (ES) [m + H] / z calculated 432, found 432.
Example 70: N- [2- (5-Isoquinolin-4-yl-1-yl-in & azol-3-yl) -3 H-benzo-imidazol-5-yl] methanesulfonamide-y *, '* • m [Anisol = Anisol] 0 (a) Intermediate substance 70a - N-. { 2- [5- Isoquinol? N-4-? L-l- (4-methoxy-benzyl) -lfl-mdazol-3-yl] -3f-benzo? M? Dazol-5-? L} -methanesulfonamide: The title compound was prepared in an 89% yield from the intermediate, Ißf * and N- (3,4-d? ammophen? l) methanesulfonamide (See Rajappa et al., Indian J. Chem. Sect. B, 19, 7, 1980, 533-535), d "d similar to the procedure used for intermediate 7c '. XH NMR (300 MHz, MeOD-d4) d 9.26 (s, ÍH) > 8.63 (s, ÍH), 8.49 (s, ÍH), 8.20 (d, ÍH, J - 7.8 Hz), 7.96 0 (d, 1H, J = 7.8 Hz), 7.72-7.81 (m, 3H), 7.58- 7.63 (m, 3H), 7.35 (d, 2H, J = 8.7 Hz), 7.16 (br s, HI), 6.89 (d, 2H, J * > 8.7 Hz), 5.76 (s, 2H), 3.75 ( s, 3H), 2.93 (s, 3H). Y. (b) EXAMPLE 70 N- [2- (5-Isoquinol-n-4-l-liy-indazol-3-yl) -3-f-benzo-m-dazol-5-l] -methanesulfonamide: 5 % -n -. ^^ t The title compound was prepared in a renßffiußnio á < 448% by PMB deprotection of the intermediate 70a analogously to the emulated procedure for example 62. 'H NMR (300 MHz, DMSO- <) d 1383 (s, ÍH), 1304 (s, ÍH) .954 (br s, ÍH), 940 (s.1H), 860 (s, 1H), i 55 X ís. ÍH), 827 (d, 1H, J = 87 Hz), 772-788 (m, 4H), 763 (dd, ÍH, J = 87.15 Hz), 754 (br s, ÍH), 746 $ ,. ÍH), 707 (d, 1H, J = 78 Hz) .291 (s, 3H) Analysis • 105 H20) C, H, N, S MS (ES) lm + H] / z calculated 455, found 455 Example 71: N-. { 2- [5- (4-met l- [3,4 'Jb? Pr? D ru.l-5-il rlfr-nda2ol-3-? L) -3f-benzo midazole-5- l] -me tanosuif onamide [Sulfur = Sulfur Anisol = Anisol " (a) Intermediate 71a -1- (4-Met? x? -benzyl) -5- (4-methyl- [3,4 '] b? p? r? dm? l-5-? l) - lfí-indazol -3-carbaldehído: *? * 262 \ S "The title compound was prepared by yielding 76% from the intermediate 19e ^ and the intermediate substance 53a in a manner similar to the * * - procedure used for the intermediate 610 »lH 5 ÍJMR (300 MHz, CDC13) d 10.28 (s, ÍH), 8.73 (dd, 2H, J =. 1.5 Hz), 8.49 (s, ÍH), 8.43 (s, ÍH), 8.31 (s, ÍH), 7.53 (d, ÍH, J - 8.7 Hz), 7.40 (dd, ÍH, J = 8.7, 1.5 Hz), 7.26-7.34 (m, 4H), 6.89 (d, 2H, J - 8.7 Hz), 5.67 (s, 2H), 3.79 (s, 3H), 2.15 (s, 3H). Analysis (C27H22N402 • 0.25 H20) C, H,. ' 0 (b) Intermediate substance 71b - 2? -. { 2- [5- (4-ethyl- [3,4 '] bipyr? Dinyl-5-yl) -1- (4-methoxy-benzyl) -I-yl-indazol-3-yl) -3-yl-benzoimida-2-yl-5- il] -methanesulfonamide: The title compound was prepared in a 96% yield from intermediate 71a 5 and hh (3, -diam? nophenyl) methanesulfonaraide in a manner similar to the procedure used for intermediate 7c '. 1 H NMR (300 MHz, MeOD-d 4) d 8.66 (dd, 2 H, J = 4.5, 1.5 Hz), ^ .52 k 1 .-, * ÍS, 2H), 8.39 (s, ÍH), 7.73 (d, ÍH, J = 8.7 Hz), 7.47-7, .65 (m, 5H), 7.35 (d, 2H, J = 8.7 Hz), 7.17 ( br s, ÍH), 6.86 (d, 2H, J = 8.7 Hz), 5.72 (s, 2H), 3.73 (s, 3H), 2.94 (s, 3tí) ', * « 2. 24 (s, 3H).
(C) Example 71 - N-. { 2- [5- (4-met? L-3, 4 /] bipyridinyl-5-yl) -lfl-indazol-3-yl) -3? -benzoimidazol-5-yl] -methanesulfonamide: ! - **? : tp, 263 x The title compound was prepared in a yield of 35% by means of the PMB deprotection of the intermediate 71b in a manner analogous to the procedure used for example 62.? 'E NMR (300 MHz, DMSO-c) δ 13.79 (s, 1H), 13.02 (s, ÍH), 9.56 (s, 1H), 8.70 (d, 2H,. - 5.7 Hz), 8.54 (s, 1H ), 8.47 (d, 2H, J - 7.8 Hz), 7.78 (d, ÍH, J * 8.7 Hz), 7.49-7.58 (m, 5H), 7.10 (d, 1H, J *? '. 7, Hz) , 2.93 (s, 3H), 2.17 (s, 3H). Analysis (C2sH21N702S • 1.45H20) C, H, N, S. MS (ES) [m + H] / z calculated 496, found fe. S '., T 10 Example 72:. { 5- [3- (1-Benzo? M? Dazol-2-yl) -lfi-mda2? L * $ -Il] -4-met l-p? R? Din-3- l} -methanol * * (a) Intermediate 72a - (5-Brom * -4-met? l-pindm-3? l) -methanol: ff »" '' 264 > ** 4 Intermediate 59a (1.5 g, 7.5 mmol) was stirred in MeOH at a temperature of 0 ° C, Sodium borohydride (850 mg, 22.5 ramol) was added and The reaction was stirred for 1 hour, the solution was diluted with ethyl acetate and the organics were washed with H 2 O and brine, dried (MgSO 4) and concentrated in vacuo, purification by chromatography on silica gel. % to 100% ethyl acetate / hexanes) yielded 1.39> g-e (92%) of intermediate 72a as a white solid XH NMR (300 MHz, CDC13) d 8.59 (s, 1H ),, 8.38 .., ** (s, ÍH), 8.43 (s, 1H), 4.75 (d, 1H, J = 5.4 Hz), 2.45 4 (s, 3H), 2.37 (t, ÍH, J = 5.4 Hz). (b) Intermediate substance 72b-. { 5- [3- (1-Benzo? M? Dazol-2-yl) -1- (4-methox? -benz? L) -lfl-indazol-5-yl] -4-met? Lp? Ridin-3 -? l} -methanol: The title compound was prepared in a *** 92% yield from intermediate 62b and intermediate 72a in a manner similar to the procedure used for intermediate 61c. 1H 'NMR (300 MHz, CDC13) d 10.48 (s, ÍH), 8.62 (s, ÍH), 8.48 (s, 1H), 8.42 (s, ÍH), 7.83 (br s, ÍH), 7.45 (d, 2H, J = 8.7 Hz), 7.21-7.32 (m, 5H), 6.83 (d, 2H, J - 8.7 Hz), 5.60, (d, 2H, J = 4.5 Hz), 4.80 (s, 2H), 3.76 (s, 3H), 2.25 (s, 3H), 1.94 (br s, ÍH).
* T X 'A.ISJ? SM = J rr * • '* .rt: *** i r *? » 265 (c) Example 72 -. { 5- [3- (1-Benzoxm? Dazol-2-yl) -l-? Nda2? L-5-? L] -4-met? L-p? R? Dm-3-? L} -methanol: The title compound was prepared in u? *, yield of 59% by means of PMB deprotection of intermediate 72b analogously to the procedure used for example 62. XH NMR (300 MHz, DMSO-dg) d *? 13.78 (s, ÍH), 13.01 (s, Í 8.39 (s, ÍH), 7.75 (dd, ÍH, "7.2 Hz), 7.50 (d, ÍH, J = 7.2 Hz), 7.44 (dd, ÍH, J = 8.4, 0 1.5 Hz), 7.15-7.23 (m, 2H), 5.29 (t, ÍH, J = 5.4 Hz), 4.64 (d, 2H, J = 5.4 Hz), 2.22 (s, 3H). Analysis (C2? H? 7N50 • 1.25H20) C, H, N. MS (ES) [m + H] / z calculated 356, found 356.
Example 73: { 5- [3- (lfí-Benzo? M? Dazol-2-? L) -lfífcinda2? J. »- 5-5 íl] -4 -met? Lp? R? D? N -3? L-met ? l} -d? met l-amma * i (a) Example 73 -. { 5- [3- (lfl-Ben # o? M? Dazol-2-yl) -lfí-ndazol-5-? L] -4 -met? Lp? R? D? N -3? -methyl } -dim til-amma: IX 268 »intermediate 59a. XH NMR (300 MHz, CDC13) d 8.50 (s, 1HJ, 8.22 (s, 1H), 2.36 (s, 3H), 2.30 (s, 3H), *. * 4 (b) Intermediate substance 75b- 3- 4 | 2- ^^ * J Benzo? Midazole-2-? L) -5- (4, 5-d? Met? Lp? Idin-3-? L) -1- (4- * methoxy-benzyl) - 1-mdazole: 1 The title compound was prepared in a 79% yield from intermediate 62b and intermediate 75a in a similar manner to > procedure used for intermediate 61c. XH 10 NMR (300 MHz, CDC13) d 10.66 (s, ÍH), 8.64 (s, ÍH), 8.40 (s, * 1H), 8.36 (s, ÍH), 7.81-7.85 (m, ÍH), 7.41- 7.49 (m, 2H), 7.Í8-7.27 (m, 5H), 6.81 (d, 2H, J = 4.5 Hz), 5.57 (s, 2H), 3.74 (s, 3H), 2.31 (s, 3H) , 2.16 (s, 3H). (c) Example 75-3- (l-Benzo-madazole-2-? l) -5- (4,5- * l * i't-? d? met? lp? r? din? 3-? l ) -Ifl-mdazole: p The title compound was prepared at a rx * yield of 37% by means of PMB deprotection of the "F * intermediate 75b in a manner similar to Example 62. 1H" NMR (300 MHz, DMSO -de) d 13.76 (s, 1H), 13.02 (s, ÍH), 842 20 (S, ÍH), 8.38 (s, ÍH), 8.31 (s, 1H), 7.74 (dd, 1H, J = 8.4, 0.6Hz), 7.51-7.69 (m, 2H), 7.44 (dd, 1H, J = 8.7, 1.8 Hz), # * 7.17-7.22 (m, 2H) 2.33 (s, 3H), 2.18 (s, 3H) . Analysis (C2? H17N5 # 1.0 269 gjerft lo 76: 3- [3- (lfí-Benzo? m? dazol-2-jl) -lfí-ifidazol-5-? J] -4- [Anisol = Anisole] (a) Intermediate 76a-3- [3- (l-Benzo? M? Da2? L-2-? L) -1- (4-methox? -benz? L) -lfl-mdazole -5-yl] -4-m-l-qu? Nol? Na: The title compound was prepared in an 86% yield from intermediate 62b and 3-bromo-4-met? Lqumol? Na ( see K on et al., Synthesis, 1976, 249) in a manner similar to the procedure used for intermediate 61c. X NMR (300 MHz, CDC13) d 10.24 (s, ÍH), 8.94 (s, 1H), 8.78 (s, ÍH), 8.25 (dd, ÍH, J = 7.8, **., 0.3Hz), 8.14 ( dd, ÍH, J = 7.8, 0.3 Hz), 7.75-7.88 (m, 2H), 7.64-7.70 (m, ÍH), 7.45-7.57 (m, 3H), 7.27-7.35 (m, 4H), 6.91 ( d, 2H, J = 6.9Hz), 5.68 (s, 2H), 3.82 (s, 3H), 2.70 (s, 3H). Analysis (C32H20N5O • 0.15 H20) C, H, N. 2 * 270 (b) Example 76- 3- [3- (l-Benzoimidazol-2-yl) -lfl-mdazol-5-yl] -4-methyl-quinoline: The title compound was prepared in * a yield of 72% by medium of PMB deprotection of intermediate 76a in a manner similar to the procedure used for example 62. lE NMR (300 MHz, DMS0-dfi) d 13.81 (s, ÍH), 13.06 (s, ÍH), 8.86 (s, ÍH), 8.55 (s, ÍH), «.23 (d, ÍH, J = 7.8 Hz), 8.09 (d, 1H, J = 7.2 Hz), 7.68-7.84 (m, 3H), 7.55-7.59 (m , 3H), 7.17-7.23 (m, 2H), 2.66 (s, 3H). Analysis (C2H? 7N5 • 0.8 H20) C, H, N. MS (ES) Im + H] / z calculated 376, found 376.
Example 77: 5- [3- (1-Benzoimidazol-2-yl) -lfl-jpdazpl-5-yl] -4-m t? L-pi £ Jdin-3-ol i NaOMe OMF MeOH 180 ° C, CO, «Cetone 654C 8 ^ / ^ O ffl (61%) N (40%) N 77» 77b n ß [Acétone = Acetone Anisol = Anisole] (a) Intermediate 77a-5-Bromo-4-me il-p? R? Dm-3-ol: 3,5-D? Bromo-4-met? Lp? R? d (Na (2.42 g, 9.64 mmol) and sodium methoxide (3.12 g, 57.8 mmol) were stirred in a mixture of DMF (8 ml) and MeOH (2 ml) in a sealed tube at 180 ° C for 24 hours. The reaction was either cooled or concentrated in vacuo. Purification by chromatography on silica gel (100% ethyl acetate) gave 1.10 g (61 *) of the intermediate 77a as a white solid. H NMR (300 MHZ, DMSO-ds) d 10.31 (s, 1H), 8.12 (s, ÍH),, 8.04 (s, ÍH), 2.21 (s, 3H). Analysis (C6H6BrNO) C, H, N. (b) Intermediate 77b-3-Bromo-5- (4-svethoxy-benzoyloxy) -4-met? L-p? R? D? Na: Intermediate 77a (1.0 g, 5.3 mmol.), Tetramethylammonium iodide (107 mg, 0.53 mmol) and potassium carbonate (1.47 g, 10.6 mmol) were stirred in acetone (30 mL). p-Methoxybenzyl (1.08 ml, 7.98% * 5 mmol) and the reaction was stirred at 55 ° C for 8 hours.The solution was diluted with ethyl acetate.The organic compounds were washed with H20 and brine, dried (MgSO 4) and concentrated in vacuo Purification by chromatography on silica gel (25% acetate 10 afforded 648 mg (40%) of the substance form of a white solid XH NMR '- 8.33 (s, ÍH) , 8.16 (s, ÍH), 7.34 (d, 2H, , í. (d, 2H, J = 8.7 Hz), 5.08 (s, 2H), 3.83 (s, 3H), 2.34 (s,: • 3H). Analysis (C? 4H? 4BrN02) C, H, N. v * fc .. 15"(c) Intermediate substance 77c- 5- [3- (14- * '* * Ben2oimida2ol-2-? L) -1- (4-methoxy-benzyl) -lfl-indazol-5-yl] -4-methyl-pyrid? N-3-ol: The title compound was prepared in an 89% from intermediate 62b. '* / * 4 20 and intermediate 77b, similar to; procedure used for intermediate 61c. XH MMR (300 MHz, CDCI3) d 10.17 (s, ÍH), 8.64 (s, ÍH), 8.29 (s, 1H), 8.25 (s, ÍH), 7.81-7.86 (m, 1H), P 274"" -4- (a > Example 78-. {5- [3- (1-Benzimddazole-2-? Ljt. * Ii? -? Ndazol-5-yl] -4-methyl-p ? nd? n-3-? l-methyl-isoprop? l-am? nat The title compound was prepared in a 28% solution from Example 72 onwards. with a procedure similar to the preparation of intermediate 68b. XH NMR (300 MHz, DMSO-dg) d 13.76 (s, 1H), 13.01 (s, 1H), 7.74 (d, 1H, (d, ÍH, J * = 7.2Hz), 7.43 (dd, ÍH, J = 8.7, 1.5Hz), 7.15-7.25 Ír i (m, 2H), 3.79 (s, 2H), 2.80-2.86 (m, ÍH), 2.27 (s, 3H), 1.07 (d, 6H, J = 6.6Hz). Analysis (C24H24N6 • 0.7H20) C, H, N. M (ES) [m + HJ / z calculated 397, found 397.
Example 79: (5-Isoquinol-n-4-? L-l-yldazol-3-? L-methylepo-p-rrol-1-yl-amine «- [Toluene = Toluene] K - * - i j Oa (aV Intermediate substance 79a-5-Isoqu? Nolm-4-yl- via 1- (2-tr? Met? Lsilan? L-ethox? Met? L) -lfl-ndazole-3-carbaldehyde: The title compound was prepared in a manner similar to the preparation of the intermediate lif. replacing by a SWL protection (see intermediate 3a) the PMB protection of the substance int rpted á "-19c .. XH NMR (300 MHz, CDC13) d 10.30 (s, 1H), 9.30 (s, Ifi),. *% 8.54 (s, 1H), 8.48 (s, 1H), 8.08 (dd, ÍH, J = 6.6, 2.4Hz) '/ * * 7.86 (dd, 1H, J = 6.6, 0.6Hz), 7.81 (dd, ÍH, J «8.7, 0.9Hz), 7.64-7.70 (m, 3H), 5.91 (s, 2H), 3.66 (t, 2H," J = 8.4Hz), 0.97 (t, 2H, J = 8.4Hz) , -0.02 (s, 9H). (b) Intermediate 79b- [S-Isoqüin lift-4-yl-l- (2-trimethylsilane-ethoxymethyl) -lJf-i »da2ol-3-yl-ethylene] -pyrrol-1-yl-amine: Intermediate 79a (400 mg, 0.99 ramol) and 1-aminopyrrole (98 rag, 1.2 mmol) were stirred with p-toluenesulfonic acid (10 mg) in toluene (6 ml) at 80 ° C for 2 hours. The solution was concentrated in vacuo and purified by chromatography on silica gel (50% ethyl acetate / hexanes) to give 410 mcf (89%) of the intermediate 79b as a yellow oil.? E NMR ( 300 MHz, CDCI3) d 9.31 (s, ÍH), 8.77 (s, ÍH), 8.58-8.60 (m, 2H), 8.09 (dd, ÍH, J = 7.3, 0.9Hz), 7.92 (d, ÍH, J = 7.8Hz), 7.76 (dd, 1H, J = 8.7, 0.9Hz), 7.63-7.70 (m, 3H), 7.19 (t, 2H, J = 2.4 z), 6.26 if "(t, 2H, J = 2.4Hz), 5.85 (s, 2H), 3.66 (t, 2H, J = 8.4Hz), 0.97 (t, 2H, J = 8.4Hz), -0.02 (s, 9H). (c) Example 79- (5-Isoquinolin-4-yl-1-yl-indazol-33-yl-methylene) -pyrrole-1-yl-amma: The title compound was prepared in a 68% yield by of the SEM checkout of the Intermediate substance 79b similar to the procedure used for example 41. XH NMR (300 MHz, DMS0-d #) d 13.82 (s, ÍH), 9.38 (s, ÍH), 9.09 (s, ÍH), 8.52 (S , ÍH), 8.41 (s, ÍH), 8.24 (dd, ÍH, J = 7.2, 1.5Hz), 7.71-7.85 (m / 4H), 7.61 (dd, ÍH, J = 8.4, 1.5Hz), 7.46 ( t, 2H, J »2.4Z), 6.15 (t, 2H, J = 2.4Hz). Analysis (C2? H15N5) C, H, N. MS (ES) [m + H] / z calculated 338, found 338. r * Example 80: 2-f5- (5-Etylamino-Nomethyl-4-methyl-pyridin-3-yl) * lJ: - indazol-3-yl] - H-benzoimidazole-4-methylamide methylamide (b) Intermediate intermediate 80b-5-Bromo-4-methyl-pyridin-3-yl-methyl) -ethyl-ethyl-ethyl ester of carbáraic acid: The intermediate 80a (850 mg, 3.7 branch) 5 was stirred in a THF solution (80 ml) and 1 N NaOH (10 ml). Di-tert-butyl dicarbonate (1.09 g, 5 mmol) was added and the reaction was stirred for 2 hours at room temperature, the solution was diluted with ethyl acetate.The organics were washed with H20 and brine. dried (MgSO 4) and concentrated in vacuo Purification by chromatography on silica gel (33% ethyl acetate / hexanes) gave 760 mg (62%) fe of the intermediate 80b as a clear oil XH NMR (300 MHz, CDC13) d 8.60 (s, ÍH),., 8.24 (s, ÍH), 4.49 (s, 2H), 3.18 (bs, 2H), 2.39 (s, 3JÍ), '' * 1.47 (s , 9H), 1.05 (t, 3H, J = 7.2 Hz)., (C) Intermediate substance Oc- Ethyl- [5- (3-butyl-phenyl-n-nazol-5-yl) -4-methyl-p ? ddin-3-yl-methyl] ethyl ester of carbamic acid: The title compound was prepared in an 85% yield from intermediate 19ie, * and intermediate 80b in a similar manner to the procedure used for intermediate 61c. * XH * NMR (300 MHz, CDC13) d 10.27 (s, ÍH), 8.38 (s, 1H), 8.35 (s-, ÍH), 8.23 (s, ÍH), 7.49 (dd, ÍH, J = 8.7, 0.6Hz) , 7.26-7.51 • * [Sulfur = Anisol Sulfur = Anisole] (a) Intermediate 81a-3-Methalosulfanyl-2-nitro-phenylamine: 3-Chloro-2-n-tro-anamma (1.0 g, 5.8 mmol) and carbonate of Potassium (880 mg, 6.4 mmol) was stirred in dry DMF (15 ml) in a sealable tube at 0 ° C. Metanetiol was bubbled through the solution for 4 minutes. The tube was sealed and the reaction was stirred at 122 ° C for 16 hours. The cooled reaction was diluted with H20 and extracted with ethyl acetate. The organic compounds were washed with brine, dried (Na2SO4) and concentrated in vacuo. Purification by chromatography on silica gel (33% • < * - * ethyl acetate / hexanes) provided 950 mg (89%) of the intermediate substance 81a as a bright red-orange solid. "H NMR (300 MHz, CDC13) d 7.21 (t, A. »? Í ~ 282 'and ÍH, J = 8.1Hz), 6.55 (d, 2H, J = 8.1Hz), 5.93 (bs, 2H), 2.42. (s, 3H). Analysis (C7H8N202S) C, H, N, S, * (b) Intermediate 81b-3-Methylsuifanyl-1-f, 1-behzeno-l, 2-diamine: f The title compound was prepared in a 96% yield from intermediate 81a in a manner similar to the hydrogenation process * - < v f 'developed for the intermediate substance 9a'. 1 H NMR (300 MHz, CDC13) d 6.93-6.97 (m, HH), 6.63-6.70 (m, 2H), 3.71 fbs, 0 4H), 2.36 (s, 3H). Analysis (C7H? 0N2S) C, H, N, S. (c) Intermediate 81c- Ethyl-. { 5- [1- (4-Ethoxy-benzyl) -3- (4-methylsulfanyl-1-yl-benzoimidazol-2-yl) -I-yl-indazol * -5-yl] -4-methyl-pyridin-3-yl-methyl } dimethyl ethyl carbamic acid ester: The title compound was prepared in an 80% yield from intermediate 81b and intermediate 80c in a manner similar to the procedure used for intermediate 7s'. XH NMR (300 MHz, CDCl 3) d 10.15 (s, 0.6H), 10.12 (s, 0. H), ft 8.64 (s, 0.4H), 8.59 (s, 0.6H), 8.47 (s, ÍH), 8.37 (s, 1H), 7.72 (d, 0.6H, J = 7.5 Hz), 7.45 (t, ÍH, J = 7.2 Hz), 7.19- '7.34 (m, 5H), 7.10 (d, 0.4H, J = 7.5 Hz), 6.83-6.89 (m, $ f) t, - 5.65 (s, 1.2H), 5.61 (s, 0.8H), 4.55 (bs, 2H), 3.78"(s, 1.8H), 3.77 (s, 1.2H), 3.26 (bs, 2H), 2.67 (s, 1.2H), 2. Sl * £ * "' (s, 1.8H), 2.24 (s, 1.2H), 2.22 (s, 1.8H), 1.49 (s, 9H) V X and ** 1. 12 (t, 3H, J = 6.9 Hz). (d) Example 81- Et? l-4-. { met? l-5- [3- (4-, t. * 4 methylsulfamyl-1-benzo-m? dazal-2-? l) -lfl-mdazol-5? l] -p? pdm-3? l-met? l} -amine: The title compound was prepared in a & amp; amp; yield of 25% by means of PMB deprotection of the * (i •, - intermediate substance 81c in a manner similar to the procedure used for example 62. 1H NMR (300 MHz, DMSO-d <; f) 13.78 (s, ÍH), 13.10 (s, ÍH), 8.48 (S, 1H), 8.44 (s, ÍH), 8.39 (s, 1H), 7.75 (d, ÍH, J = 8.7 Hz), 7.44 (d, 1H, J * 8.7, 1.8 Hz), 7.30 (d, 1H, J - 7.8 Hz), 7.19 (t, ÍH, J * 7.8 rf Hz), 6.99 (d, 1H, J = 7.2 Hz) , 3.82 (s, 2H), 2.66 (q, 2H, J j. • = 7.2 Hz), 2.56 (s, 3H), 2.28 (s, 3H), 1.08 (t, 3H, J »7.2 * £ Hz) . Analysis (C24H24N6S • 1.5H20) C, H, N, S. MS (83 [m + H] / z calculated 429, found 429. 1" Example 82: N-. { 2- [5- (5-Et? Lam? Nomet? L-4-roetjlrtir? D? N-3-il) - * "£ -. 284 * [Sulfur = Anisol Sulfur = Anisole] (a) Intermediate substance 'ß2a- 2J 2,3-dian Í < "* faith .1) -trace acetate: The title compound was prepared in a 98% yield from N- (2-ammo-3-n-tro-phenyl) -acetamide (See Harvey et al., J. Chem. Soc. Perk. Trans. 1, 1988, 1939-1944) similar to the hydrogenation of the intermediate substance 9a '. 1 H NMR (300 MHz, CDC13) d 9.04 (a, ÍH), 6.35-6.49 (m, 3H), 4.38 (bs, 4H), 2.00 (s, 3H). (b) Intermediate substance 82b- { 5- [3- (4-Acet? lam? no-lfl-benzoim? dazol- 2-? L) -1- (4-methox? -benz? L) - fí - indazol-5-? 1] -4-methyl-p? r? d n -3? -methyl} ethyl ester of carbamic acid: The title compound was prepared in 65% yield from intermediate 82a and intermediate 80c in a manner similar to the procedure used for intermediate 7c 'lH NMR (300 MHz, CDC13) d 12.35 (bs, 1H), 10.80 (bs, ÍH), 7.90-8.85 (m, 4H), 6.76-7.46 (m, 8H), 5.60 (bs, 2H), 4.51 (bs) , 2H), 3.78 (s, 3H), 3.61 (bs, 2H), 3.19 (bs, 3H), 1.74 (bs, 12H), 1.18 (bs, 3H). MS (ES) [m + H] / z calculated 660, found 660. i? p $? ' (c) Example 82- N- (2- [5- (5-Etylaminomethyl-4-yl] et? l-pyridin-3-yl) -lfl-indazol-3? l] -lfi-benzo3.m Dazol-4-yl.) Acetamide: The title compound was prepared in wñ 6% yield by means of PMB deprotection of intermediate * 82b in a manner similar to the procedure used for example 62. XE NMR (300 MHz, MeOD-d4) d 8. 64 '(s, ÍH), 8.58 (s, ÍH), 8.45 (s, ÍH), 7.82 (d, 1H, J *, 8.4Hz), 7.57 (d, ÍH, J = 7.2Hz), 7.50 (d, 2H, J = 7.2Hz), 10 7.34 (t, 1H, J = 8.4Hz), 4.44 (s, 2H), 3.27 (q, 2H, J * 7.5Hz), 2.43 (s, 3H), 2.22 (s, 3H), 1.41 (t, 3H, J = 7.5Hz). MS (ES) [m + H] / z calculated 440, found 440.
Example 83: 5- (2,6-D? Fluorophenyl) -3-f n l-lfl-indazole • * • k 5 83 -tm .. = 18b H2, Pd / C EtOAc * 99% 85 84 With a synthetic method similar to that used for 5 intermediate substance 18c, the hydrogenation of 5-n? Tro-3- (2-p? Rrol? L) -1 H -mdazole 18b 'on 10% palladium in * carbon afforded 5-ammo-3- (2-pyrrolidol) -Iff-indazole 84 (99%) in the form of a beige solid: lE NMR (DMSO-d6) d 6.13 (def, 1H, J = 2.4, 2.6 Hz), 6.49 (dd, ÍH, J = 1.5, 2.4 0 Hz), 6.76 (dd, ÍH, J = 1.5, 2.6 Hz), 6.79 (dd, ÍH, J = 2.1, 8.9 Hz), 7.03 (d, ÍH, J = 2.1 Hz), 7.22 (d, 1H, J = 8.9 Hz), 11.16 (s, ÍH), 12.45 (s, 1H). Analysis (C? .Hx0N4 »0.2 ethyl acetate) C, H, N. * »--'-. , 5 The title compound was prepared in an 81% yield from Example 72 and isobutylamines with a procedure similar to the preparation of intermediate 68b. -H NMR (300 MHz, DMSO-de) d 13.77 (s, ÍH), 13.02 (s, ÍH), 8.50 (s, ÍH), 8.42 (s, 1H), 8.37 (s, ÍH), 7.74 (d, ÍH, J = 8.7Hz), 7.68 (d, 1H, J * 7.5Hz), 7.50 V. (d, ÍH, J = 7.5Hz), 7.43 (dd, ÍH, ^ 8.7,1.5Hz), 7.14-7.24 * (m, 2H), 3.85 (s, 2H), 2.49 (bs, 2H), 2.27 (s, 3H), 1 * 73- f .'i 1.79 (m, ÍH), 0.90 (d, 6H, J = 6.6Hz). Analysis (C2SH26N6 * 0.3 H20) C, H, N. MS (ES) [m + H] / z calculated 411, found yf 411.
Ejefnplp 88? . { 5- [3- (lfl »Benzo? M? Dazol-2 ~ jl) -Üffi-indazol-S-il] - * 4-met l-pj¿, id? N-3-il-met l} -benz l-amine (a) Example 88-. { 5- [3- (1-Benzo [mu] midazole-2? -1) -1? E »mdazol-5? 1] -4-met? L-p? Pdm-3? -methyl} -bengyl-amine The title compound was prepared in a yield of 73% from example 72 and benzylamine in a manner similar to intermediate 68b. XH NMR (300 MHz, DMS0-d6) d 13.77 (s, ÍH), 13.02 (s, ÍH), 8.51 (s, ÍH) "- 8.42 (s, ÍH), 8.38 (s, ÍH), 7.74 (d , 1H, J * 8.7 Hz), 7.6 # (d, 1H, J = 7.5 Hz), 7.50 (d, ÍH, J = 7.5 Hz) », 7.16-7.44 (m, 8H), 3.90 (bs, 4H) , 2.23 (s, 3H). Analysis (C28H2N6 • 1.2H20) C, H, N. MS (ES) [m + H] / z calculated 445, found 445.
Example 89: 2- ( { 5- [3- (lfí-Ben2? M? Da2 l-2-? L) -lfí-m aa £ l * - l] -4 - ?? et lp? r? d? n-3-? l-met l.}. -am? no-ethanol (a) In n nal substance) -dial flow: A solution of 2, 3-d? Met? L-7-n? Thromdol (Acros Organics, 13.55 g, 71.24 mmol) in diechloromethane (1.0 L) was cooled to an internal temperature of -60 ° C and a greenish yellow. Argon was bubbled through the solution for one hour, causing the color change to yellow. Dimethylsulfide (10.5 rol, ») 142. 5 mmol) and stirring continued at -60 ° C for 1.5. s hours. After warming to room temperature, the solution was concentrated in vacuo to ~ 200 ml, washed with water (2 x 50 ml), dried over magnesium sulfate, filtered, concentrated and purified by gel * chromatography. of silica (50 to 100% ethyl acetate efn hexanes), which yielded compound 90a (11.85 g, * 75%) in the form of an orange solid. Rf = 0.36 (75% ethyl acetate / hexanes); XH NMR (DMSO-d6) d 2.02 (s, 3H), 2.52 (s, 3H), 7.52 (t, ÍH, J = 7.9 Hz), 8.00 (dd 1H, J = 7.9, 1.5 Hz), 8.05 (dd, ÍH, J = 8.1, 1.5 Hz), 10. 32 (s, 1H). Analisis (C? OH? 0N2O4 • 0 .4 H20) C, H, N. (b) Intermediate 9Qb-l- (2 -! &fnino-3-nitro-j enyl) -ethanon: Concentrated hydrochloric acid (40 ml) was added to a solution of 90a (4.00 g, 18.0 mmol) in absolute ethanol (80 ml) and water (40 ml). The mixture was heated to reflux (internal temperature of 87 ° C) for 1 hour. After cooling to room temperature, a saturated aqueous solution of sodium bicarbonate was added to bring the pH to 8. The solution was extracted with ethyl acetate (2 x 200 * ml). The combined organic extracts were dried magnesium sulfate sulfate, filtered, concentrated and purified by chromatography on silica gel (20 to 70% ethyl acetate in hexanes) to give compound 90b (2.67 g, 82%) as of a yellow solid. Rf = 0.45 (50% ethyl acetate / hexanes); XH NMR (DMSOefe >? D, 2.62 (s, 3H), 6.74 (t, ÍH, J = 8.1 Hz), 8.31 (m 2H), 8.85 (br s, 2H) Analysis (C8H8N203) C, H, N. (c) Intermediate 90c-l- (2,3-Diamino-pheny1) -ethanone: With a synthetic method similar to the synthesis of 9a, the hydrogenation of 90b (2.00 g, 11.1 mmol) in ethanol provided Compound 90c (1.54 g, 92%) in the form of bright yellow crystals Rl = O.34 (50% ethyl acetate / hexanes); X NMR (DMSO-d6 d '2.47 (s, 3H), 4.75 (br s, 2H), 6.40 (dd, ÍH, J «7.5, ' * 295 8. 1 Hz), 6.69 (dd, 1H, J = 7.5, 1.3 Hz), 6.79 (br s, 2H), 7.10 (dd, ÍH, J = 8.1, 1.3 Hz). Analysis (CßHioNjO) C, H, N. (d) Intermediate substance 90d - l-. { 3*. { 5- Isoquinol? N-4-? Ll- (4-methox? -benz? L) -lfí-mda2? L -3? L] -lfí ~ * ben2om? Da2? L-4-? L) -ethanone : In a manner similar to the 19hr synthesis, aldehyde 19f (2.02 g, 5.13 mmol) and diam 90c (771 mg, 5.13 mmol) were condensed in the presence of sulfur to provide 90d (1.83 g, 68%) as a bright yellow solid. Rf = 0.19 (75% 4-ethyl acetate / exanos); XH NMR (DMSO-d6) [Some * • * peaks were doubled due to the isome izaoion 'tautomeric] d 2.72 and 2.87 (2 brs, 3H as a whole), # ^ "3.71 (s, 3H), 5.85 (s) , 2H), 6.93 (d, 2H, J = 8.7 Ha), 7.34 (m, 3H), 7.75 (m, 5H), 8.07 (ra, 2H), 8.25 (d, ÍH, J * 7.5 Hz), 8.56 and 8.80 (2 br, 2H as a whole), ft.38, (s, ÍH), 11.83 (s, ÍH), 13.53 (s, ÍH). (e) Intermediate substance 90e - (l- { 2 - [5- ", 'Isoquinolin-4-? II- (4-methox? -benz? L) -lfl-? Ndazol-3? L] -lfi-benzoam? Dazol-4-íl} -ethyl) -methyl-amine: A solution of methylamine in methanol (2.0 M, 3.02 mL, 6.04 mmol) was added to the ketone 90d (527.8 mg, 1.01 mmol) at room temperature, followed by hydrochloric acid (4.0 mL). in dioxane, 0.504 ml, 2.02 mmol), ^ m ta ol (6.0 ml) and sodium cycloborohydride (38.0 mg, 0.605 mmol). The suspension was stirred at RT 4 * > Environment for 23 hours, but nirif ^ 'tj • > The reaction was carried out by TLC analysis, anhydrous THF (10 ml) was added to increase the solubility and the stirring was continued for 70 hours.The mixture was partitioned between ethyl acetate and a saturated aqueous sodium bicarbonate solution. Organic phase was dried over magnesium sulfateit was filtered, concentrated and purified by chromatography on silica gel (1: 20: 200 B0H - aqueous: tanol: d? chloromethane), which yielded compound 90e (275.0 rag, 51%) as a foam yellow. Rf = 0.09 (1: 20: 400 aqueous NH 4 OH: ethanol: dichloromethane); ? E NMR (CD30D) d 1.53 (d, 3H, J = 6.8 Hz), 2.24 (s, 3H), 3.75 (s, 3H), 4.28 (q, ÍH, J = 6 * 8 Hz), 5.45 (s, ÍH), 5.77 (s, 2H), 6.89 (d, 2H, J «= 8.7 'Hz), 7.22 (m, 2H), 7.34 (d, 2H, J = 8.7 Hz), 7.52 (d , ÍH, J = 7.9 Hz), 7.61 (dd, 1H, J = 8.7, 1.5 JÍZ), 7.76 (m, 3H), 7.99 (d, ÍH, J = 8.3 Hz), 8.20 (dd, ÍH, J = 7.2, 0 1.7 Hz), 8.48 (s, ÍH), 8.70 (s, ÍH), 9.27 (s, 1H). * Analysis (C34H3oN60 »1.0 H20) C, H, N. 'i and ^ (f) Example 90 -. { 1- [2- (5-Isoqui-olin ~ 4-? L-lf? -4-? Ndazol-3-? L) -lfi-benzo? M? Dazol-4 ~ l] -ethyl} - * netil-am? na: A solution of 90e (179.7 mg, 0.334 mmol), * 5 trifluoromethanesulfonic acid (0.84 ml) and acid i 297 trifluoroacetic (3.34 ml) was stirred at 50 ° C for 2 hours. The solution was then added dropwise to a rapidly stirred mixture of concentrated aqueous NH40H (10 ml) and ethyl acetate (30 ml). Extraction and purification similar to that of Example 33, provided the compound * 90 * in the form of a whitish solid * * (140.9 mg). Although this material appeared pure by HPLC and XH NMR analysis, the analysis of elements revealed significant impurities. The impure material was dissolved in ethyl acetate (50 ml) and washed with water (10 ml), saturated aqueous sodium bicarbonate solution (10 ml) and saturated aqueous sodium chloride solution (10 ml). The organic layer was dried over magnesium sulfate, filtered and concentrated to give compound 90 (49.4 mg, 35%) as a white solid: XH NMR (CD3OD) d 1.71 (d, 3H, J = 6.8 Hz), 2.45 (s, 3H), 4.66 (q, 1H, J = 7.0 Hz), 7.24 (d, 1H, J *, • 7.5 Hz), 7.32 (t, ÍH, J = 7.7 Hz) , 7.61 (dd, ÍH, J * 7.9, 1.0 Hz), 7.67 (dd, ÍH, J = 8.5, 1.5 Hz), 7.82 (m, 2T 3H), 8.03 (d, ÍH, J = 8.3 Hz), 8.24 (d, 1H, J «7.5 Hz), 8.53 (s, ÍH), 8.71 (s, ÍH), 9.30 (s, ÍH) Analysis Yes (C26H22N6'0.4 CH2C12) C, H, N. after 20 minutes with the addition of ethylenediaminetetraacetic acid (EDTA) up to 250 mM. The phosphorylated substrate was then captured on a nitrocellulose or phosphocellulose membrane using a 96-well filament manifold and the unincorporated radioactivity was removed%. by repeated washing with 0.85% phosphoric acid. Radioactivity was quantified by exposing the dried membranes to an optical scanner with phosphorus storage techniques (Phosphorimager). The apparent values of K ± were measured by assays of the enzymatic activity in the presence of P different concentrations of inhibitory compounds and r subtraction of background radioactivity measured in the absence of the enzyme. The inhibition data were entered into an equation for competitive inhibition using the Kaleidagraph application (Synergy Software) or entered into an equation for competitive inhibition for strong bonds using the KmeTic software (BioKm, Ltd.).
Inhibition of kinase activity of the retinffelastoma kinase CDK4 / Cyclamen D A complex of human CDK4 and cyclin D3, or a complex of human CDK4 and (1-264) cyclma D3 genetically truncated, was purified using traditional chromatographic biochemical techniques with insect cells what # * • were co-infected with the corresponding baculovirus expression vectors (See for example, Meijer and Kim, "Chemical Inhibitors of Cyclin-Dependent Kinases," Metho? ls in Enzymol,., vol.283 (1997), pp. 113-128 .). The enzyme complex (5 or 50 nM) was tested with 0.3-0.5 μg of purified recombinant protein fragment of retinoblastoma (Rb) as a substrate. The Rb engineering fragment (residues 386-928 of the native retinoblastoma protein, 62.3 kDa) contains the majority of the phosphorylation sites found in the native 106-kDa protein, and also an indicator of histidine residues to facilitate , purification. ' The phosphorylated substrate Rb was captured by microfiltration on a nitrocellulose membrane and quantified using a phosphorimager as described above. For the measurement of strong ligation inhibitors, the concentration of the enzyme complex was decreased to 5 nM, and the duration of the test was extended to 60 minutes, during which the time-dependent formation of the product was linear.
Inhibition of the kinase activity of CDK2 / Cyclin A gibenblase A CDK2 was purified using published methodology (Rosenblatt et al., "Purification and Crystallization of Human Cyclin-dependent Kinase 2," J. Mol. Biol. , vol 230, 1993, pp. 1317-1319) from cells ** < • .303 of insects infected with baculovirus expression vectors. Cyclin A was purified from *, J E. coli cells expressing recombinant cyclin A of full length, a truncated cyclin A construct was generated by limited proteolysis and purified as previously described (Jeffrey et al., "Mechanism of CDK activation revealed by the structure of a cyclin A-CDK2 complex," Nature, vol. 376 (July 27, 1995), p. 313-320). A complex of CDK2 and proteolyzed cyclin A was prepared and purified by gel filtration. The substrate for this erisa, I * J was the same Rb substrate fragment used for the / sac of CDK4 and the CDK2 / cyclin A and r CDK4 / cyclin D3 assay methodology was essentially the same, except that CDK2 was present in values of 150 nM or 5 nM. The values of Kx were measured as described above. > - The stimulation of cell proliferation by growth factors, such as VEGF and others, depends on the induction of autophosphorylation of each of the tyrosine kinases of their respective receptors. Therefore, the ability of an inhibitor of * ¿. , «Protem-qumasa to stop the cell proliferation induced by these growth factors is directly correlated with its ability to block the autophosphorylation of the receptor. For m dir ^ *? < * 304 inhibitory activity of protein kinase of 1 $ 4ÉÍ - ». compounds, the following constructions were used ..
Construction for the VEGF-R2 test This construction determines the capacity of * tt > - • test compound to inhibit dß activity. the tyrosine kinase. A construction (VEGF-R2? 50) of the domain,. Cytosolic receptor 2 of the human endothelial vascular growth factor (VEGF-R2) with absence of the 50 central residues of the 68 residues of the kinase insertion domain, was expressed in a baculovirus / cell system of > •; * insect. Of the 1356 full-length residues VEGF-R2, VEGF-R2? 50 contains residues 806-939 and 990-1171, as well as a point mutation (E990V) within the domain and insertion of the kinase relative to VEGF-R2 of wild type. The autophosphorylation of the purified construct was carried out by incubation of the enzyme at a concentration of I *. μM in the presence of 3 mM ATP and 40 mM MgCl2 in Hepes 100 jraM, *, ", - pH 7.5, containing 5% glycerol and 5 mM DTT, at 4 ° C for 2 hours After autophosphorylation, it is demonstrated that construction possesses essentially catalytic activity - equivalent to the construction of the wild-type autophosphorylated kinase domain (See Parast et al., Biochemistry, 37, 16788-16801 (1998).
- - £ *. * Construction for the ens, of QHK1 The full length human CHK1 (FL-CHK1) with His and C terminal indicators was expressed using the baculovirus / insect cell system. It contains 6 histidine residues (6 x His indicator) in the C terminal of the human CHKl of amino acid 476. The protein was purified by standard chromatographic techniques. 10 VEGF-R2 assay Coupled spectrophotometric assay (FLVK-P) The production of ADP from ATP that encapsulated the phosphoryl transfer was coupled to the oxidation of NADH using phosphoenolpyruvate (PEP) and a system having p-pyruvate kinase (PK) and lactic dehydrogenase (LDH). The oxidation of NADH was monitored by monitoring the decrease in absorbance at 340 nm (e34o = 6.22 cm "1 mM" 1) using a Beckman DU 650 spectrophotometer. The conditions "of the assay for the phosphorylated compound VEGF-R2? 50 2 & (indicated as FLVK-P in the tables below) were the. following: 1 mM PEP; NADH 250 μM; 50 units of LDH / i ^ l; 2 * PK / ml units; 5 mM DTT; poly (E4Y?) 5.1 mM; ATP 1 mM f "Í '^ 25 mM MgCl2 in 200 mM Hepes, pH 7.5. The df-assay conditions for the non-phosphorylated compound VEGF-R2? 50 (indicated 25 as FLVK in the tables) were the following: PEP1 mM; t NADH 250 μM; 50 units of LDH / mL; 20 units of PK / mL; 5 mM DTT; poly (E4Y?) 20 mM; 3 mM ATP; MgCl260 pM and 2 aM MnCl2 in 200 mM Hepes, pH 7.5. The assays were started with 5 to 40 nM enzyme. The Kx values were determined by measuring the enzymatic activity in the presence of varied conditions of the test compounds. The data was r- * analyzed using the Enzyme Kinetic and Kaleidagraph software.
ELISA assay The formation of phosphogastrin was monitored using biotinylated gastrin peptide (1-17) as a substrate. The "*" biotinylated phosphogastrin was immobilized using 96-well microtiter plates coated with streptavidite, followed by detection using anti-phosphotyrosine antibodies conjugated with horseradish peroxidase The activity of horseradish peroxidase was monitored using p-salt of 2, 2 '-azmo-di- [3-ethylbenzathiazoline sulfonate (6)] (ABTS) .'Typical assay solutions contain: 2 μM biotinylated gastrin peptide, 5 mM DTT, 20 μM ATP, MgCl226 μg, and M Cl2 2 mM in 200 mM Hepes, pH 7.5 The assay was started with 0.8 nM phosphorylated VEGF-R2 50. The activity of horseradish peroxidase was evaluated using 10 mM ABTS The reaction of horseradish peroxidase was stopped the * acid addition (H2S04), followed by the absorbance reading at 405 nm.The Kx values were determined by "í the measurement of the enzymatic activity in the presence of variable concentrations of the test compounds. The data were analyzed using the software Enzyme Kínetíc and ¡i t X Kaleidagraph.
CHKl assay The production of ADP from ATP that accompanies •.
The transfer of phosphoryl to the synthetic peptide substrate Syntide-2 (PLARTLSVAGLPGKK) was coupled to the oxidation of NADH using phosphoenolpyruvate (PEP) through the action of pyruvate kinase (PK) and lactic dehydrogemase (LDH). Oxidation of NADH was monitored by monitoring the decrease in absorbance at 340 nm (€ 340 = 6.22 cm "1 mM" 1) using a HP8432 spectrophotometer Typical reaction solutions contained: 4 mN PEP, NADH 0.15 mM, 28 units of LDH / ml, 16 units of PK / ml, 3 mM DTT, 0.125 mM Syntide-2, 0.15 mM ATP, 25 mM MgCl2 in 50 mM TRIS, pH 7.5 and 400 mM NaCl. 10 nM of FL-CHK1 The Ki values were determined by measuring the initial enzymatic activity in the presence of varying concentrations of the test compounds.The * data were analyzed using the Enzyme Kínetic and Kaleidagraph software. • ste. * 308 Inhibition of the phosphorylated FGF receptor and LCK tyrosine kinase activity The cloning, expression and purification of the tyrosine kinase dointin cytosolic FGFR1 (amino acids 436 5 766) containing three substituted amino acids (L457V, 488A «and C584S) was conducted as previously described ', (Mohammadi, M., Schlessinger, J., &Hubbard, SR (199 * 6) • * Cell 86, 577-587). This domain was expressed in Sf9 insect cells using a baculovirus expression vector, and the protein was purified using standard * ß techniques. LCK tyrosine kinase was expressed in insect cells - cpmo deletion of N terminals beginning at amino acid * 223 until the end of the protein at amino acid 509. The N-terminus of the protein also had two substitutions of 15 amino acids, P223M and C 224D. The kinases were purified using conventional chromatographic methods. The tyrosine kinase activity was measured using a coupled continuous spectrophotometric assay, in which the production of phosphorylated substrate poly (Glu, T? 4; 1) and 0 'ADP is coupled to the transfer of a phosphoenolpyruvate phosphate to ADP, catalyzed by pyruvate kinase, with the generation of pyruvate and the regeneration of ATP. The production of pyruvate in turn is coupled with the reduction of pyruvate to form lactate, catalyzed by lactic 5-dehydrogenase, with the concomitant conversion of i *** $ • -3í $ 8W NADH to NAD +. The loss of NADH is monitored by measuring the absorbance at 340 nm (see for example ^ Technikova-Dobrova et al., "Spectrophotometric determination of functional characteristics of protein. csupled enzymatic assay, "FEBS Letters, vol 292 (1991)," pp. 69-72) Enzyme activity was measured in the presence of 200 mM HEPES (pH 7.5), 2 mM phosphoenolpyruvate, 0.3 mM NADH, 20 mM MgCl2, 100 μM ATP, 5 mM DTT, poly JGlu, Tir) 4: 1, 5.1 or 25 mM for the P-FGF or P-LCK assays, respectively, and 15 units / ml of each of pyruvate kinase and lactic dehydrogenase. The phosphorylated kinase of the phosphorylated FGF receptor was present in 100 nM and the kinase phosphorylated LCK in 50 nM. The tests were carried out under conditions of initial proportions at 37 ° C and the proportions were corrected according to the measurements of background proportions in the absence of the enzyme. The percentage of inhibition was calculated in relation to the enzyme? control assayed in the presence of 2% (v / v) DMSO. The results are shown in Table 1.
Coupled spectrophotometric assay (FAK) Tyrosine kinase assays were monitored using a Beckman DU 650 spectrophotometer. The ADP production was coupled to the oxidation of NADH using phosphoenolpyruvate (PEP) by the action of the * á H Í 310 pyruvate qumasa (PK) and lactic dehydrogenase (LDH). The oxidation of NADH was monitored by following the lafß: decrease in absorbance at 340 nm (e34c = 6-22 cm "1 mM"). Typical reaction solutions contain: 1 mM PEP, 250 μM NA H, 50 LDH units / ml, 20 PK units / ml, DTT 5,. tm mM, in 200 mM Hepes, pH 7.5 and vanablef concentrations of poly of cdFGFRl. The results of the tests carried out with the compounds, which include the specific examples described above, are indicated below in Table I. Except where otherwise indicated in a particular line, the units and tests used are as indicated in Applicable column of the table.
Table I: Kx with kinases * x c 311 1t. «? * 'P tea i > . * • saw" * ( Note: NT = not tested f Inhibition of cell growth: assessment of cytotoxicity Inhibition of cell growth is iftidio * f. using the tetrazoyl salt assay, which is based on the ability of viable cells to reduce 3- (4,5-d? met? lt? azol-2-? l) -2, 5- [2H] -bromide from diphenyltetranolium (MTT) to formazan (Mossman, Journal of Immunological Methods, vol 65 (1983), pp. 55-58). The product formazan purple insoluble in water was then detected spectrophotometrically. The HCT 116 cell line was incubated - ^ in 96-well plates. The cells were placed in pI Gas in the appropriate medium at a volume of 135 μl / well in the McCoy 5A Medium. The plates were incubated for four hours before adding the inhibitor compounds. Different concentrations of compounds were added *. ** i * ^ • inhibitors in 0.5% (v / v) of dimethyl? Ulphoxide 6.5 'μl / well) and the cells were incubated at 37 ° C (5% C02) > for four to six days (depending on the cell type). At the end *, of the incubation, MTT was added to a concent ^ afdi? N *? final 0.2 mg / ml and the cells were incubated for 4. «. • * more hours at 37 ° c After the centrifugation of the, '< 4 plates and the removal of the medium, the absorbance of the forraazan (solubilized in dimethylsulfoxide) was measured at 540 nm. The concentration of the inhibitor compound causing 50% of the growth inhibition was determined from the linear portion of a semilogarithmic graph of%? -inhibitor concentration as a function of the -percentage "of inhibition. All results were compared with r * control cells treated only with dimethylsulfoxide 0.5% (v / v).
Table II X - 321 4 * s incorporates a suitable dosage unit form pax'a • administration by injection.
Oral Composition To prepare a pharmaceutical composition for oral administration, 100 mg of a compound or fß '' Formula I or II is mixed with 750 mg of lactose. The mixture is incorporated into an oral dosage unit, such as capsules of A »hard gelatin, suitable for oral administration. * It should be understood that the foregoing description is of an exemplary and explanatory nature, and prefers to illustrate the invention and its preferential materializations. Therefore, the scope of the invention must be understood in its definition, not by the previous description, but by the following claims and equivalent sug.
It is noted that in relation to this date, the best known method for the applicant to carry out the aforementioned invention, is the conventional one for the manufacture of the objects or products to which the • same refers.

Claims (1)

  1. '323"wherein R 4 is H or a lower alkyl, and X is an alkyl, aryl, heteroaryl, carbocycle or substituted on substituted heterocycle, or the pharmaceutically acceptable salt of a compound of Formula I, or a pharmaceutically active prodrug or metabolite of a compound of Formula I, or a pharmaceutically metabolite salt. 2. A computer acceptable, a prodrug, or active according to claim 1, characterized in that: Ri is X «^ v ** > , where R4 is hydrogen; or where Y is CH or N and R3 is H, or an alkyl group, I. aryl, carbocycle, hetero 25 or a pharmaceutically acceptable salt, pharmaceutically active prodrug or metabolite, or a pharmaceutically acceptable salt of a prodrug or metabolite. 32 » of R is H or a lower alkyl, and X is a substituted or unsubstituted heteroaryl, carbocycle or heterocycle; or a pharmaceutically acceptable salt of, ün -f * compound of Formula I; a prodrug or a pharmaceutically active metabolite of a compound of Formula I, or a pharmaceutically acceptable salt of a prodrug or metabolite; and a pharmaceutically acceptable carrier. 5. A method of treatment of diseases; 0 or disorders mediated by the inhibition of a complex of Xt¿ 4 * 'kinase, which includes the administration to the patient that requires it *, a cell cycle control agent *. selected from the group consisting of a compound - \ where R 4 is H or a lower alkyl, and X is a alkyl, aryl, heteroaryl, carbocycle or heterocycle cl group; substituted or unsubstituted; and R2 is an alkyl, apl, heteroaryl, carbocycle or heterocycle group, substituted or unsubstituted, or where R 4 is H or a lower alkyl, and X is a substituted or unsubstituted, substituted or unsubstituted heteroaryl group; or a pharmaceutically acceptable salt of a compound of Formula I; a prodrug or a pharmaceutically active metabolite of a compound of Formula I, or a pharmaceutically acceptable salt of a prodrug or metabolite. 6. A pharmaceutical composition comprising: (a) a therapeutically effective amount of a compound represented by Formula I: -fr. 'replaced; or -C (0) -R 10 where > ? or is: H; an optionally substituted alkyl, aryl, alkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl group, or OR100 or NR100R110, wherein R100 and R110 are each independently H or a ~ 1 alkyl, alkenyl group , alkynyl, cycloalkyl, heterocycloalkyl, aryl or heteroaryl, - and optionally substituted, R 2 is H or alkyl; R7 and R8 are each independently H or a replaced; where R1, R4, R5, R6 and R7 are each H, Rs is unsubstituted phenyl; or a pharmaceutically acceptable salt, prodrug, active metabolite or solvate of the compound. 10. A method according to Claim 1, wherein the protein kinase receptor is a CDK, 10 VEGF or CHK1 complex. 11. A pharmaceutical composition, which Wherein Ri is hydrogen or an alkyl, aryl ", 4 * heteroaryl, carbocycle or heterocycle, substituted or unsubstituted group, or v * where R 4 is H or a lower alkyl, and X is an alkyl, aryl, heteroaryl, carbocycle or heterocycle group, "substituted or unsubstituted R 2 is a group or substituted heterocycle H > where R 4 is H or a lower alkyl, and X is an aryl, heteroaryl, carbocycle or heterocycle group, substituted or unsubstituted; or X 'a pharmaceutically acceptable salt of a compound of Formula I; a prodrug or a pharmaceutically active 1% metabolite of a compound of Formula 1, or uncontrolled, which includes administering to the patient that requires a therapeutically effective amount of a compound represented by Formula I: 25 -. 25 - * J «J«? • tí »• i,; * where Ri is hydrogen or an alkyl, aryl, * i? teteroaryl, carbocycle or heterocycle, substituted or substituted group, or W * > where R 4 is H or a lower alkyl, and X is an aryl, heteroaryl, carbocycle or heterocycle, substituted or substituted nd group; or a pharmaceutically acceptable salt of a compound of Formula I; a prodrug or a pharmaceutically active metabolite of a compound of Formula I, or pharmaceutically active of a compound of Formula II or 4 ^ iina pharmaceutically acceptable salt of the prodrug "or imetabolite. 14. A pharmaceutically acceptable compound, a pharmaceutically acceptable salt, a prodrug, or metabolite 10 active according to claim 13, characterized in that R'i is claim 13, characterized in that R'2 is a group - 19, A compound in accordance with. the claim 13, characterized in that R '2 is a group' * $ "x 25 substituted or unsubstituted of the formula V-s 20. A compound according to claim 12, characterized in that it has the structure: or a pharmaceutically acceptable salt thereof. 21. A compound according to claim 15, characterized in that the agent that inhibits PARP is 22 A compound selected from the group consisting of ** ¡S * ^ 25 ** ^ g 25 »%, *** ' 25 5 25"#V 25 'F? Vtii 9- X 349 S « , * Or a pharmaceutically acceptable salt thereof, a prodrug or a pharmaceutically active metabolite, or a pharmaceutically acceptable salt or X + prodrug of a prodrug or an inetabolite 23. A pharmaceutical composition for the treatment of a disease associated with cell proliferation. uncontrolled comprising: i) a compound as claimed in the * Claim 13 or a pharmaceutically acceptable salt, prodrug or metabolite p pharmaceutically active or a pharmaceutically acceptable salt of a metabolite? prodigal, and 25 ii. a pharmaceutically acceptable carrier. * S i > Y*? 24. A method of disorder associated with uncontrollable, comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of Claim 13, or a pharmaceutically acceptable salt of a compound of Formula-II; or a prodrug, or metab < A pharmaceutically active marker of a compound of Formula II, or a pharmaceutically acceptable salt of the prodrug or metabolite. 25. A compound according to claim 1, characterized in that Ri is ^ S * "where R'2 is * 25 ? ~ 4 5
MXPA02007058A 2000-01-18 2001-01-18 Indazole compounds, pharmaceutical compositions, and methods for mediating or inhibiting cell proliferation. MXPA02007058A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US17648400P 2000-01-18 2000-01-18
PCT/US2001/001477 WO2001053268A2 (en) 2000-01-18 2001-01-18 Indazole compounds, pharmaceutical compositions, and their use for mediating or inhibiting cell proliferation

Publications (1)

Publication Number Publication Date
MXPA02007058A true MXPA02007058A (en) 2003-01-28

Family

ID=22644536

Family Applications (1)

Application Number Title Priority Date Filing Date
MXPA02007058A MXPA02007058A (en) 2000-01-18 2001-01-18 Indazole compounds, pharmaceutical compositions, and methods for mediating or inhibiting cell proliferation.

Country Status (39)

Country Link
US (4) US6555539B2 (en)
EP (1) EP1250326A2 (en)
JP (1) JP2003520273A (en)
KR (1) KR20020073505A (en)
CN (1) CN1394205A (en)
AP (1) AP1609A (en)
AR (1) AR032438A1 (en)
AU (1) AU785013B2 (en)
BG (1) BG107011A (en)
BR (1) BR0107783A (en)
CA (1) CA2388885A1 (en)
CO (1) CO5280070A1 (en)
CR (1) CR6630A (en)
DO (1) DOP2001000120A (en)
DZ (1) DZ3301A1 (en)
EA (1) EA200200768A1 (en)
EE (1) EE200200398A (en)
GE (1) GEP20043363B (en)
GT (1) GT200100009A (en)
HN (1) HN2001000007A (en)
HR (1) HRP20020675A2 (en)
HU (1) HUP0203965A3 (en)
IL (1) IL150730A0 (en)
IS (1) IS6474A (en)
MA (1) MA27589A1 (en)
MX (1) MXPA02007058A (en)
MY (1) MY136604A (en)
NO (1) NO20022117L (en)
NZ (1) NZ518531A (en)
OA (1) OA12160A (en)
PA (1) PA8509901A1 (en)
PE (1) PE20011334A1 (en)
PL (1) PL357590A1 (en)
SK (1) SK10052002A3 (en)
SV (1) SV2002000293A (en)
UA (1) UA75880C2 (en)
WO (1) WO2001053268A2 (en)
YU (1) YU54202A (en)
ZA (1) ZA200203040B (en)

Families Citing this family (196)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020150921A1 (en) * 1996-02-09 2002-10-17 Francis Barany Detection of nucleic acid sequence differences using the ligase detection reaction with addressable arrays
CZ301750B6 (en) 1999-12-24 2010-06-09 Aventis Pharma Limited Bicyclic pyrrole derivative, its use in the preparation of a medicament, pharmaceutical composition in which the derivative is comprised and for use in therapy
YU54202A (en) * 2000-01-18 2006-01-16 Agouron Pharmaceuticals Inc. Indazole compounds,pharmaceutical compositions,and methods for mediating or inhibiting cell proliferation
US6897231B2 (en) * 2000-07-31 2005-05-24 Signal Pharmaceuticals, Inc. Indazole derivatives as JNK inhibitors and compositions and methods related thereto
US20050009876A1 (en) * 2000-07-31 2005-01-13 Bhagwat Shripad S. Indazole compounds, compositions thereof and methods of treatment therewith
US7211594B2 (en) 2000-07-31 2007-05-01 Signal Pharmaceuticals, Llc Indazole compounds and compositions thereof as JNK inhibitors and for the treatment of diseases associated therewith
WO2002016348A1 (en) * 2000-08-09 2002-02-28 Astrazeneca Ab Antiangiogenic bicyclic derivatives
ES2302106T3 (en) 2000-09-11 2008-07-01 Novartis Vaccines And Diagnostics, Inc. PROCEDURE FOR PREPARATION OF BENCIMIDAZOL-2-IL QUINOLINE DERIVATIVES.
DE10046029A1 (en) 2000-09-18 2002-03-28 Bayer Ag indazoles
US6982274B2 (en) 2001-04-16 2006-01-03 Eisai Co., Ltd. 1H-indazole compound
GB0115109D0 (en) 2001-06-21 2001-08-15 Aventis Pharma Ltd Chemical compounds
US7642278B2 (en) 2001-07-03 2010-01-05 Novartis Vaccines And Diagnostics, Inc. Indazole benzimidazole compounds
EP1401831A1 (en) 2001-07-03 2004-03-31 Chiron Corporation Indazole benzimidazole compounds as tyrosine and serine/threonine kinase inhibitors
FR2827861B1 (en) 2001-07-27 2004-04-02 Aventis Pharma Sa DERIVATIVES OF INDAZOLES OR INDOLES, THEIR USE IN HUMAN MEDICINE AND MORE PARTICULARLY IN CANCEROLOGY
US20040077702A1 (en) * 2001-09-14 2004-04-22 Wen-Mei Fu Treatment of nuerodegenerative diseases
EP1432416B1 (en) 2001-09-26 2011-01-19 Pfizer Italia S.r.l. Aminoindazole derivatives active as kinase inhibitors, process for their preparation and pharmaceutical compositions containing them
JP4637481B2 (en) 2001-10-19 2011-02-23 オーソ−マクニール・フアーマシユーチカル・インコーポレーテツド 2-phenylbenzimidazole and imidazo- [4,5] -pyridine as CDS1 / CHK2 inhibitors and adjuvants for chemotherapy or radiotherapy in cancer treatment
FR2831536A1 (en) * 2001-10-26 2003-05-02 Aventis Pharma Sa NOVEL BENZIMIDAZOLE DERIVATIVES, PROCESS FOR THEIR PREPARATION, THEIR USE AS MEDICAMENTS, PHARMACEUTICAL COMPOSITIONS AND NOVEL USE IN PARTICULAR AS KDR INHIBITORS
FR2831537B1 (en) * 2001-10-26 2008-02-29 Aventis Pharma Sa NOVEL BENZIMIDAZOLE DERIVATIVES, PROCESS FOR THE PREPARATION THEREOF, THEIR USE AS MEDICAMENTS, PHARMACEUTICAL COMPOSITIONS AND THE USE THEREOF
IL161576A0 (en) * 2001-10-26 2004-09-27 Aventis Pharma Inc Benzimidazoles and analogues and their use as protein kinases inhibitors
US6897208B2 (en) 2001-10-26 2005-05-24 Aventis Pharmaceuticals Inc. Benzimidazoles
US7074805B2 (en) * 2002-02-20 2006-07-11 Abbott Laboratories Fused azabicyclic compounds that inhibit vanilloid receptor subtype 1 (VR1) receptor
US6867320B2 (en) * 2002-02-21 2005-03-15 Asahi Kasei Pharma Corporation Substituted phenylalkanoic acid derivatives and use thereof
NZ535349A (en) * 2002-03-08 2007-01-26 Signal Pharm Inc JNK inhibitors with chemotherapeutic agents in a combination therapy for treating or preventing cancer and other proliferative disorders in refractory patients in particular
FR2836914B1 (en) 2002-03-11 2008-03-14 Aventis Pharma Sa SUBSTITUTED INDAZOLES, COMPOSITIONS CONTAINING SAME, METHOD OF MANUFACTURE AND USE
US7166293B2 (en) 2002-03-29 2007-01-23 Carlsbad Technology, Inc. Angiogenesis inhibitors
US20050119278A1 (en) * 2002-05-16 2005-06-02 Che-Ming Teng Anti-angiogenesis methods
MXPA04011851A (en) * 2002-05-30 2005-03-31 Celgene Corp Methods of using jnk or mkk inhibitors to modulate cell differentiation and to treat myeloproliferative disorders and myelodysplastic syndromes.
IL164209A0 (en) * 2002-05-31 2005-12-18 Eisai Co Ltd Pyrazole derivatives and pharmaceutical compositions containing the same
JP2004075673A (en) * 2002-06-19 2004-03-11 Mitsubishi Chemicals Corp Compound and organic electroluminescent element using the same
GB0218625D0 (en) * 2002-08-10 2002-09-18 Astex Technology Ltd Pharmaceutical compounds
EP1545515A1 (en) 2002-08-12 2005-06-29 Sugen, Inc. 3-pyrrolyl-pyridopyrazoles and 3-pyrrolyl-indazoles as novel kinase inhibitors
US20040087642A1 (en) * 2002-10-24 2004-05-06 Zeldis Jerome B. Methods of using and compositions comprising a JNK inhibitor for the treatment, prevention, management and/or modification of pain
EP1582211A1 (en) * 2002-12-03 2005-10-05 Kyowa Hakko Kogyo Co., Ltd. Jnk inhibitor
EA008501B1 (en) 2002-12-19 2007-06-29 Пфайзер Инк. 2-(1h-indazol-6-ylamino)-benzamide compounds as protein kinases inhibitors useful for the treatment of ophtalmic diseases
US20050019366A1 (en) * 2002-12-31 2005-01-27 Zeldis Jerome B. Drug-coated stents and methods of use therefor
US6933311B2 (en) * 2003-02-11 2005-08-23 Abbott Laboratories Fused azabicyclic compounds that inhibit vanilloid receptor subtype 1 (VR1) receptor
BRPI0407618A (en) * 2003-02-21 2006-02-21 Pfizer cycloalkyl-substituted amino thiazole derivatives containing n and pharmaceutical compositions for inhibiting cell proliferation and methods for their use
DE602004029145D1 (en) 2003-02-27 2010-10-28 Palau Pharma Sa Pyrazolopyridine DERIVATIVES
ES2214150B1 (en) * 2003-02-27 2005-12-01 J. URIACH &amp; CIA S.A. "NEW DERIVATIVES OF PIRAZOLOPIRIDINAS".
DE602004006852D1 (en) 2003-04-17 2007-07-19 Janssen Pharmaceutica Nv 2-PHENYL-BENZIMIDAZOLE AND 2-PHENYL-IMIDAZO-4,5'-PYRIDINE DERIVATIVES AS CHECKPOINT-KINASE-CDS1 (CHK2) INHIBITOR FOR THE TREATMENT OF CANCER
FR2854159B1 (en) * 2003-04-25 2008-01-11 Aventis Pharma Sa NOVEL INDOLE DERIVATIVES, THEIR PREPARATION AS MEDICAMENTS, PHARMACEUTICAL COMPOSITIONS AND IN PARTICULAR AS KDR INHIBITORS
US7015233B2 (en) 2003-06-12 2006-03-21 Abbott Laboratories Fused compounds that inhibit vanilloid subtype 1 (VR1) receptor
TWI372050B (en) 2003-07-03 2012-09-11 Astex Therapeutics Ltd (morpholin-4-ylmethyl-1h-benzimidazol-2-yl)-1h-pyrazoles
ITRM20030355A1 (en) * 2003-07-18 2005-01-19 Sigma Tau Ind Farmaceuti COMPOUNDS OF CYTOTOXIC ACTIVITIES COMBRETASTATINE DERIVATIVES.
KR101204247B1 (en) 2003-07-22 2012-11-22 아스텍스 테라퓨틱스 리미티드 3,4-disubstituted 1h-pyrazole compounds and their use as cyclin dependent kinases cdk and glycogen synthase kinase-3 gsk-3 modulators
CA2518951A1 (en) * 2003-07-30 2005-02-10 Kyowa Hakko Kogyo Co., Ltd. Protein kinase inhibitors
EP1652842B1 (en) * 2003-07-30 2012-04-11 Kyowa Hakko Kirin Co., Ltd. Indazole derivatives
US7008953B2 (en) * 2003-07-30 2006-03-07 Agouron Pharmaceuticals, Inc. 3, 5 Disubstituted indazole compounds, pharmaceutical compositions, and methods for mediating or inhibiting cell proliferation
US20050090529A1 (en) * 2003-07-31 2005-04-28 Pfizer Inc 3,5 Disubstituted indazole compounds with nitrogen-bearing 5-membered heterocycles, pharmaceutical compositions, and methods for mediating or inhibiting cell proliferation
US7511145B2 (en) * 2003-08-01 2009-03-31 Genelabs Technologies, Inc. Bicyclic heteroaryl derivatives
US20050113576A1 (en) * 2003-08-05 2005-05-26 Chih-Hung Lee Fused azabicyclic compounds that inhibit vanilloid receptor subtype 1 (VR1) receptor
WO2005014554A1 (en) * 2003-08-08 2005-02-17 Astex Therapeutics Limited 1h-indazole-3-carboxamide compounds as mapkap kinase modulators
EP1660427A4 (en) 2003-08-14 2006-12-20 Asahi Kasei Pharma Corp Substituted arylalkanoic acid derivative and use thereof
JP2005089457A (en) * 2003-09-03 2005-04-07 Yung Shin Pharmaceutical Industry Co Ltd Pharmaceutical composition for promoting bone growth or inhibiting bone resorption
US6984652B2 (en) * 2003-09-05 2006-01-10 Warner-Lambert Company Llc Gyrase inhibitors
MY144339A (en) * 2003-09-08 2011-08-29 Aventis Pharma Inc Thienopyrazoles
TWI344961B (en) * 2003-10-15 2011-07-11 Ube Industries Novel indazole derivative
BRPI0416266A (en) * 2003-11-06 2007-01-09 Celgene Corp method for treating, preventing and / or controlling an asbestos-related disease or disorder in a patient
EP2762475A1 (en) 2003-11-07 2014-08-06 Novartis Vaccines and Diagnostics, Inc. Pharmaceutically acceptable salts of quinolinone compounds and their medical use
US20060179711A1 (en) * 2003-11-17 2006-08-17 Aerogrow International, Inc. Devices and methods for growing plants
EP2065383A1 (en) 2003-11-19 2009-06-03 Signal Pharmaceuticals, Inc. Indazole compounds and methods of use thereof as protein kinase inhibitors
US7488817B2 (en) * 2004-02-02 2009-02-10 The Trustees Of The University Of Pennsylvania Metal complex protein kinase inhibitors
WO2005094823A1 (en) * 2004-03-30 2005-10-13 Kyowa Hakko Kogyo Co., Ltd. Flt-3 INHIBITOR
BRPI0509580A (en) * 2004-03-30 2007-11-27 Pfizer Prod Inc signal transduction inhibitor combinations
GB0409080D0 (en) * 2004-04-23 2004-05-26 Biofocus Discovery Ltd Compounds which interact with protein kinases
AU2005266494B2 (en) * 2004-07-27 2009-09-10 Novartis Ag Inhibitors of Hsp90
US20070054916A1 (en) * 2004-10-01 2007-03-08 Amgen Inc. Aryl nitrogen-containing bicyclic compounds and methods of use
FR2876584B1 (en) * 2004-10-15 2007-04-13 Centre Nat Rech Scient Cnrse USE OF KENPAULLONE FOR THE PRODUCTION OF MEDICAMENTS FOR THE TREATMENT OF MUCOVISCIDOSIS AND DISEASES ASSOCIATED WITH A DEFECT OF ADDRESSING PROTEINS IN CELLS
US20060116519A1 (en) * 2004-11-17 2006-06-01 Agouron Pharmaceuticals, Inc. Synthesis of 5-bromo-4-methyl-pyridin-3-ylmethyl)-ethyl-carbamic acid tert-butyl ester
WO2006054143A1 (en) * 2004-11-17 2006-05-26 Pfizer Inc. Polymorphs of {5-[3-(4,6-difluoro-1h-benzoimidazol-2-yl)-1h-indazol-5-yl]-4-methyl-pyridin-3-ylmethyl}-ethyl-amine
CA2588558A1 (en) * 2004-11-23 2006-06-01 Celgene Corporation Jnk inhibitors for treatment of cns injury
AU2005321091B2 (en) 2004-12-30 2012-04-12 Astex Therapeutics Limited Pyrazole compounds that modulate the activity of CDK, GSK and Aurora kinases
US20080161251A1 (en) 2005-01-21 2008-07-03 Astex Therapeutics Limited Pharmaceutical Compounds
JP5475235B2 (en) 2005-01-21 2014-04-16 アステックス・セラピューティクス・リミテッド Pharmaceutical compounds
CA2596527C (en) 2005-01-27 2013-02-12 Kyowa Hakko Kogyo Co., Ltd. Igf-1r inhibitors
GT200600042A (en) 2005-02-10 2006-09-27 Aventis Pharma Inc BIS ARILO AND HETEROARILO COMPOUNDS REPLACED AS SELECTIVE ANTAGONISTS OF 5HT2A
US20070004777A1 (en) * 2005-03-23 2007-01-04 Bhagwat Shripad S Methods for treating or preventing acute myelogenous leukemia
PT1869020E (en) 2005-03-29 2010-12-23 Icos Corp Heteroaryl urea derivatives useful for inhibiting chk1
JP2008540345A (en) * 2005-04-29 2008-11-20 セルジーン・コーポレーション Solid form of 1- (5- (1H-1,2,4-triazol-5-yl) (1H-indazol-3-yl))-3- (2-piperidylethoxy) benzene
ATE530533T1 (en) 2005-05-17 2011-11-15 Novartis Ag METHOD FOR SYNTHESIS OF HETEROCYCLIC COMPOUNDS
WO2006130673A1 (en) 2005-05-31 2006-12-07 Janssen Pharmaceutica, N.V. 3-benzoimidazolyl-pyrazolopyridines useful in treating kinase disorders
US7470787B2 (en) 2005-07-11 2008-12-30 Aerie Pharmaceuticals, Inc. Isoquinoline compounds
WO2007058626A1 (en) * 2005-11-16 2007-05-24 S*Bio Pte Ltd Indazole compounds
PT1957074E (en) 2005-11-29 2014-06-25 Novartis Ag Formulations of quinolinones
KR20070077468A (en) * 2006-01-23 2007-07-26 크리스탈지노믹스(주) Imidazopyridine derivatives inhibiting protein kinase activity, method for the preparation thereof and pharmaceutical composition containing same
AU2007213452A1 (en) * 2006-02-10 2007-08-16 Biomarin Iga Limited Treatment of Duchenne muscular dystrophy
JP2009530337A (en) * 2006-03-23 2009-08-27 エフ.ホフマン−ラ ロシュ アーゲー Substituted indazole derivatives, process for producing the same, and use thereof as a drug
CN101484427A (en) 2006-06-30 2009-07-15 协和发酵麒麟株式会社 Abl kinase inhibitor
US20090209537A1 (en) * 2006-06-30 2009-08-20 Kyowa Hakko Kirin Co., Ltd. Aurora inhibitors
JPWO2008020606A1 (en) * 2006-08-16 2010-01-07 協和発酵キリン株式会社 Angiogenesis inhibitor
CA2661898A1 (en) * 2006-08-25 2008-02-28 Abbott Laboratories Indazole derivatives that inhibit trpv1 and uses thereof
CL2007002617A1 (en) 2006-09-11 2008-05-16 Sanofi Aventis COMPOUNDS DERIVED FROM PIRROLO [2,3-B] PIRAZIN-6-ILO; PHARMACEUTICAL COMPOSITION THAT INCLUDES SUCH COMPOUNDS; AND ITS USE TO TREAT INFLAMMATION OF THE ARTICULATIONS, Rheumatoid Arthritis, TUMORS, LYMPHOMA OF THE CELLS OF THE MANTO.
CA2664335C (en) 2006-09-20 2014-12-02 Boehringer Ingelheim International Gmbh Rho kinase inhibitors
EP2073803B1 (en) 2006-10-12 2018-09-19 Astex Therapeutics Limited Pharmaceutical combinations
WO2008044045A1 (en) 2006-10-12 2008-04-17 Astex Therapeutics Limited Pharmaceutical combinations
US8071779B2 (en) 2006-12-18 2011-12-06 Inspire Pharmaceuticals, Inc. Cytoskeletal active rho kinase inhibitor compounds, composition and use
US20100022517A1 (en) * 2006-12-18 2010-01-28 Richards Lori A Ophthalmic formulation of rho kinase inhibitor compound
JP2010513557A (en) * 2006-12-20 2010-04-30 アボット・ラボラトリーズ N- (5,6,7,8-tetrahydronaphthalen-1-yl) urea derivatives and related compounds as TRPV1 vanilloid receptor antagonists for the treatment of pain
US8455513B2 (en) 2007-01-10 2013-06-04 Aerie Pharmaceuticals, Inc. 6-aminoisoquinoline compounds
WO2008106594A2 (en) * 2007-02-28 2008-09-04 The Arizona Board Of Regents, On Behalf Of The University Of Arizona Inhibitors of nucleophosmin (npm) and methods for inducing apoptosis
CN101622015A (en) 2007-03-05 2010-01-06 协和发酵麒麟株式会社 Pharmaceutical composition
US8648069B2 (en) 2007-06-08 2014-02-11 Abbvie Inc. 5-substituted indazoles as kinase inhibitors
CN101790526A (en) * 2007-06-08 2010-07-28 雅培制药有限公司 5-heteroaryl substituted indazoles as kinase inhibitors
EP2214675B1 (en) 2007-10-25 2013-11-20 Genentech, Inc. Process for making thienopyrimidine compounds
US8455514B2 (en) 2008-01-17 2013-06-04 Aerie Pharmaceuticals, Inc. 6-and 7-amino isoquinoline compounds and methods for making and using the same
EP2323989A4 (en) * 2008-03-20 2011-06-22 Abbott Lab Methods for making central nervous system agents that are trpv1 antagonists
KR101269393B1 (en) 2008-04-28 2013-05-29 아사히 가세이 파마 가부시키가이샤 Phenylpropionic acid derivative and use thereof
US8106039B2 (en) * 2008-04-30 2012-01-31 The Trustees Of The University Of Pennsylvania Metal complex phosphatidyl-inositol-3-kinase inhibitors
US8450344B2 (en) 2008-07-25 2013-05-28 Aerie Pharmaceuticals, Inc. Beta- and gamma-amino-isoquinoline amide compounds and substituted benzamide compounds
ES2465971T3 (en) 2009-04-06 2014-06-09 University Health Network Kinase inhibitors and method to treat cancer with them
ES2553827T3 (en) 2009-05-01 2015-12-14 Aerie Pharmaceuticals, Inc. Dual mechanism inhibitors for the treatment of disease
JP2013501792A (en) * 2009-08-10 2013-01-17 サミュメッド リミテッド ライアビリティ カンパニー Indazole and its therapeutic use as WNT / B-catenin signaling pathway inhibitor
DK2987487T3 (en) * 2009-08-10 2020-12-07 Samumed Llc INDAZOLINE INHIBITORS OF THE WNT SIGNAL ROAD AND ITS THERAPEUTIC USES
UY33001A (en) 2009-11-06 2011-05-31 Boehringer Ingelheim Int ARYL AND HETEROARILCARBONYL HIVAHYDROINDENOPIRIDINE AND OCTAHYDROBENZOQUINOLINE DERIVATIVES
CA2986631C (en) 2009-12-21 2020-06-02 Samumed, Llc 1h-pyrazolo[3,4-.beta.]pyridines and thereapeutic uses thereof
TWI516262B (en) 2010-04-06 2016-01-11 健康網路大學 Kinase inhibitors and method of treating cancer with same
WO2012020725A1 (en) * 2010-08-10 2012-02-16 塩野義製薬株式会社 Heterocyclic derivative having npy y5 receptor antagonism
ES2879314T3 (en) 2010-08-18 2021-11-22 Biosplice Therapeutics Inc Dketones and hydroxyketones as activators of the catenin signaling pathway
TWI537258B (en) 2010-11-05 2016-06-11 百靈佳殷格翰國際股份有限公司 Aryl-and heteroarylcarbonyl derivatives of hexahydroindenopyridine and octahydrobenzoquinoline
CA2819373A1 (en) 2010-12-09 2012-06-14 Amgen Inc. Bicyclic compounds as pim inhibitors
WO2012129338A1 (en) 2011-03-22 2012-09-27 Amgen Inc. Azole compounds as pim inhibitors
JP2014509660A (en) * 2011-04-01 2014-04-21 ユニバーシティ・オブ・ユタ・リサーチ・ファウンデイション Substituted 3- (1H-benzo {d} imidazol-2-yl) -1H-indazole analogs as inhibitors of PDK1 kinase
BR112014006127A2 (en) 2011-09-14 2017-04-04 Samumed Llc indazol-3-carboxamides and their use as inhibitors of the wnt / b-catenin signaling pathway
WO2013063321A1 (en) 2011-10-25 2013-05-02 The General Hospital Corporation Wnt/b-catenin inhibitors and methods of use
PH12017500997A1 (en) 2012-04-04 2018-02-19 Samumed Llc Indazole inhibitors of the wnt signal pathway and therapeutic uses thereof
CN104271133B (en) 2012-05-04 2017-10-13 萨穆梅德有限公司 1H pyrazolos [3,4 b] pyridine and its treatment use
WO2014041518A1 (en) * 2012-09-14 2014-03-20 Glenmark Pharmaceuticals S.A. Thienopyrrole derivatives as itk inhibitors
EP2943198B1 (en) 2013-01-08 2019-07-17 Samumed, LLC 3-(benzoimidazol-2-yl)-indazole inhibitors of the wnt signaling pathway and therapeutic uses thereof
CN104370889A (en) * 2013-01-24 2015-02-25 韩冰 Intraocular tension reducing compound and preparation method and use thereof
JP6401189B2 (en) 2013-02-22 2018-10-03 サミュメッド リミテッド ライアビリティ カンパニー Γ-diketones as activators of Wnt / β-catenin signaling pathway
WO2014134774A1 (en) 2013-03-04 2014-09-12 Merck Sharp & Dohme Corp. Compounds inhibiting leucine-rich repeat kinase enzyme activity
WO2014134776A1 (en) 2013-03-04 2014-09-12 Merck Sharp & Dohme Corp. Compounds inhibiting leucine-rich repeat kinase enzyme activity
US9440952B2 (en) 2013-03-04 2016-09-13 Merck Sharp & Dohme Corp. Compounds inhibiting leucine-rich repeat kinase enzyme activity
WO2014134772A1 (en) * 2013-03-04 2014-09-12 Merck Sharp & Dohme Corp. Compounds inhibiting leucine-rich repeat kinase enzyme activity
CN109528721B (en) 2013-03-15 2021-10-01 爱瑞制药公司 Combination therapy
EP3027605B1 (en) 2013-07-31 2017-11-08 Council of Scientific & Industrial Research Novel indazole compounds and a process for the preparation thereof
KR102195494B1 (en) 2013-10-18 2020-12-28 유니버시티 헬스 네트워크 Treatment for pancreatic cancer
CN106687454B8 (en) 2014-07-24 2019-08-30 贝达医药公司 2H- indazole derivative and its medical application as cell cycle protein dependent kinase (CDK) inhibitor
EP3206686B1 (en) 2014-08-20 2019-10-09 Samumed, LLC Gamma-diketones for treatment and prevention of aging skin and wrinkles
WO2016040180A1 (en) 2014-09-08 2016-03-17 Samumed, Llc 3-(1h-benzo[d]imidazol-2-yl)-1h-pyrazolo[3,4-c]pyridine and therapeutic uses thereof
WO2016040182A1 (en) 2014-09-08 2016-03-17 Samumed, Llc 2-(1h-indazol-3-yl)-1h-imidazo[4,5-c]pyridine and therapeutic uses thereof
WO2016040188A1 (en) 2014-09-08 2016-03-17 Samumed, Llc 3-(3h-imidazo[4,5-c]pyridin-2-yl)-1h-pyrazolo[3,4-c]pyridine and therapeutic uses thereof
WO2016040181A1 (en) 2014-09-08 2016-03-17 Samumed, Llc 3-(1h-imidazo[4,5-c]pyridin-2-yl)-1h-pyrazolo[3,4-c]pyridine and therapeutic uses thereof
WO2016040184A1 (en) 2014-09-08 2016-03-17 Samumed, Llc 3-(3h-imidazo[4,5-b]pyridin-2-yl)-1h-pyrazolo[3,4-c]pyridine and therapeutic uses thereof
WO2016040193A1 (en) 2014-09-08 2016-03-17 Samumed, Llc 3-(1h-imidazo[4,5-c]pyridin-2-yl)-1h-pyrazolo[3,4-b]pyridine and therapeutic uses thereof
WO2016040185A1 (en) 2014-09-08 2016-03-17 Samumed, Llc 2-(1h-indazol-3-yl)-3h-imidazo[4,5-b]pyridine and therapeutic uses thereof
WO2016040190A1 (en) 2014-09-08 2016-03-17 Samumed, Llc 3-(3h-imidazo[4,5-b]pyridin-2-yl)-1h-pyrazolo[3,4-b]pyridine and therapeutic uses thereof
GB201506658D0 (en) 2015-04-20 2015-06-03 Cellcentric Ltd Pharmaceutical compounds
GB201506660D0 (en) 2015-04-20 2015-06-03 Cellcentric Ltd Pharmaceutical compounds
GB201511382D0 (en) 2015-06-29 2015-08-12 Imp Innovations Ltd Novel compounds and their use in therapy
WO2017024004A1 (en) 2015-08-03 2017-02-09 Samumed, Llc. 3-(1h-pyrrolo[2,3-b]pyridin-2-yl)-1h-pyrazolo[4,3-b]pyridines and therapeutic uses thereof
US10166218B2 (en) 2015-08-03 2019-01-01 Samumed, Llc 3-(1H-indol-2-yl)-1H-pyrazolo[3,4-C]pyridines and therapeutic uses thereof
US10383861B2 (en) 2015-08-03 2019-08-20 Sammumed, LLC 3-(1H-pyrrolo[2,3-C]pyridin-2-yl)-1H-pyrazolo[3,4-C]pyridines and therapeutic uses thereof
US10285983B2 (en) 2015-08-03 2019-05-14 Samumed, Llc 3-(1H-pyrrolo[2,3-B]pyridin-2-yl)-1H-pyrazolo[3,4-B] pyridines and therapeutic uses thereof
US10285982B2 (en) 2015-08-03 2019-05-14 Samumed, Llc 3-(1H-pyrrolo[2,3-B]pyridin-2-yl)-1H-pyrazolo[3,4-C]pyridines and therapeutic uses thereof
US10226448B2 (en) 2015-08-03 2019-03-12 Samumed, Llc 3-(1H-pyrrolo[3,2-C]pyridin-2-yl)-1H-pyrazolo[3,4-B]pyridines and therapeutic uses thereof
WO2017024015A1 (en) 2015-08-03 2017-02-09 Samumed, Llc. 3-(3h-imidazo[4,5-b]pyridin-2-yl)-1h-pyrazolo[4,3-b]pyridines and therapeutic uses thereof
US10463651B2 (en) 2015-08-03 2019-11-05 Samumed, Llc 3-(1H-pyrrolo[3,2-C]pyridin-2-YL)-1H-indazoles and therapeutic uses thereof
US10206908B2 (en) 2015-08-03 2019-02-19 Samumed, Llc 3-(1H-pyrrolo[3,2-C]pyridin-2-YL)-1H-pyrazolo[3,4-C]pyridines and therapeutic uses thereof
WO2017023993A1 (en) 2015-08-03 2017-02-09 Samumed, Llc. 3-(1h-indol-2-yl)-1h-pyrazolo[4,3-b]pyridines and therapeutic uses thereof
US10231956B2 (en) 2015-08-03 2019-03-19 Samumed, Llc 3-(1H-pyrrolo[3,2-C]pyridin-2-YL)-1 H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
US10195185B2 (en) 2015-08-03 2019-02-05 Samumed, Llc 3-(1H-imidazo[4,5-C]pyridin-2-yl)-1H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
WO2017023986A1 (en) 2015-08-03 2017-02-09 Samumed, Llc 3-(1h-indol-2-yl)-1h-indazoles and therapeutic uses thereof
US10350199B2 (en) 2015-08-03 2019-07-16 Samumed, Llc 3-(1h-pyrrolo[2,3-b]pyridin-2-yl)-1h-indazoles and therapeutic uses thereof
WO2017023989A1 (en) 2015-08-03 2017-02-09 Samumed, Llc. 3-(1h-benzo[d]imidazol-2-yl)-1h-pyrazolo[4,3-b]pyridines and therapeutic uses thereof
US10519169B2 (en) 2015-08-03 2019-12-31 Samumed, Llc 3-(1H-pyrrolo[2,3-C]pyridin-2-yl)-1 H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
WO2017023988A1 (en) 2015-08-03 2017-02-09 Samumed, Llc. 3-(3h-imidazo[4,5-c]pyridin-2-yl)-1h-pyrazolo[4,3-b]pyridines and therapeutic uses thereof
CN107922349B (en) * 2015-08-07 2021-05-07 哈尔滨珍宝制药有限公司 Vinyl compounds as FGFR and VEGFR inhibitors
WO2017079759A1 (en) 2015-11-06 2017-05-11 Samumed, Llc 2-(1h-indazol-3-yl)-3h-imidazo[4,5-c]pyridines and their anti-inflammatory uses thereof
US9643927B1 (en) 2015-11-17 2017-05-09 Aerie Pharmaceuticals, Inc. Process for the preparation of kinase inhibitors and intermediates thereof
JP6832946B2 (en) 2015-11-17 2021-02-24 アエリエ ファーマシューティカルズ インコーポレイテッド How to prepare kinase inhibitors and their intermediates
AR108325A1 (en) 2016-04-27 2018-08-08 Samumed Llc ISOQUINOLIN-3-IL CARBOXAMIDS AND PREPARATION AND USE OF THE SAME
AR108326A1 (en) 2016-04-27 2018-08-08 Samumed Llc ISOQUINOLIN-3-IL CARBOXAMIDS AND PREPARATION AND USE OF THE SAME
CN109476660B (en) 2016-06-01 2023-04-04 拜斯丽治疗有限公司 Preparation method of N- (5- (3- (7- (3-fluorophenyl) -3H-imidazo [4,5-C ] pyridine-2-yl) -1H-indazol-5-yl) pyridine-3-yl) -3-methylbutyramide
US11078192B2 (en) 2016-07-05 2021-08-03 Guangzhou Maxinovel Pharmaceuticals Co., Ltd. Aromatic acetylene or aromatic ethylene compound, intermediate, preparation method, pharmaceutical composition and use thereof
WO2018013430A2 (en) 2016-07-12 2018-01-18 Arisan Therapeutics Inc. Heterocyclic compounds for the treatment of arenavirus infection
EP3506890A1 (en) 2016-08-31 2019-07-10 Aerie Pharmaceuticals, Inc. Ophthalmic compositions
CA3041291A1 (en) * 2016-10-21 2018-04-26 Samumed, Llc Methods of using indazole-3-carboxamides and their use as wnt/b-catenin signaling pathway inhibitors
US10758523B2 (en) 2016-11-07 2020-09-01 Samumed, Llc Single-dose, ready-to-use injectable formulations
SG11201908179UA (en) 2017-03-31 2019-10-30 Aerie Pharmaceuticals Inc Aryl cyclopropyl-amino-isoquinolinyl amide compounds
US10604514B2 (en) 2017-10-19 2020-03-31 Samumed, Llc 6-(5-membered heteroaryl)isoquinolin-3-yl carboxamides and preparation and use thereof
WO2019084497A1 (en) 2017-10-27 2019-05-02 Samumed, Llc 6-(6-membered heteroaryl & aryl)isoquinolin-3-yl carboxamides and preparation and use thereof
US10413537B2 (en) 2017-10-27 2019-09-17 Samumed, Llc 6-(5-membered heteroaryl)isoquinolin-3-yl-(5-membered heteroaryl) carboxamides and preparation and use thereof
US10703748B2 (en) 2017-10-31 2020-07-07 Samumed, Llc Diazanaphthalen-3-yl carboxamides and preparation and use thereof
JP7293560B2 (en) 2017-12-29 2023-06-20 グアンジョウ マキシノベル ファーマシューティカルズ カンパニー リミテッド Aromatic vinyl or aromatic ethyl derivative, its production method, intermediate, drug composition and use
BR112020017087A2 (en) 2018-02-23 2020-12-22 Samumed, Llc INDAZOL-3-CARBOXAMIDES REPLACED 5-HETEROARILLA AND PREPARATION AND USE OF THE SAME
US10759799B2 (en) 2018-06-15 2020-09-01 Samumed, Llc Indazole containing macrocycles and therapeutic uses thereof
WO2020056345A1 (en) 2018-09-14 2020-03-19 Aerie Pharmaceuticals, Inc. Aryl cyclopropyl-amino-isoquinolinyl amide compounds
WO2020150545A1 (en) 2019-01-17 2020-07-23 Samumed, Llc Pyrazole derivatives as modulators of the wnt/b-catenin signaling pathway
WO2021088859A1 (en) * 2019-11-06 2021-05-14 暨南大学 Indazole compound, pharmaceutical composition of same, and applications thereof
WO2021125229A1 (en) * 2019-12-17 2021-06-24 富士フイルム株式会社 Indazole compound or salt thereof, and pharmaceutical composition
WO2021121420A1 (en) * 2019-12-20 2021-06-24 江苏凯迪恩医药科技有限公司 Benzopyrazole compound and intermediate, preparation method, and application thereof
WO2021168341A1 (en) 2020-02-21 2021-08-26 Samumed, Llc Solid forms of 1-(5-(3-(7-(3-fluorophenyl)-3h-imidazo[4,5-c]pyridin-2-yl)-1h-pyrazolo[3,4-b]pyridin-5-yl)pyridin-3-yl)-n,n-dimethylmethanamine
CN112724134B (en) * 2021-01-14 2022-04-01 复旦大学 Azaindazole bipyridine derivative myeloid cell proliferation inhibitor, preparation method and application thereof in pharmacy
WO2023240138A1 (en) * 2022-06-08 2023-12-14 Blossomhill Therapeutics, Inc. Indazole containing macrocycles and their use

Family Cites Families (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1376600A (en) 1971-02-15 1974-12-04 Agfa Gevaert Photographic developer compositions
CH575397A5 (en) 1971-11-09 1976-05-14 Basf Ag
US3994890A (en) * 1974-01-31 1976-11-30 Chugai Seiyaku Kabushiki Kaisha 1-Aminoalkyl, 3-phenyl indazoles
FR2265739B1 (en) 1974-03-29 1976-12-17 Ugine Kuhlmann
JPS5615287A (en) 1979-07-16 1981-02-14 Chugai Pharmaceut Co Ltd Pyrazoloindazole derivative and its preparation
EP0104029B1 (en) 1982-09-17 1988-08-24 Kudelski S.A. Control system for an electric motor
JPS59228248A (en) 1983-06-08 1984-12-21 Konishiroku Photo Ind Co Ltd Posi-posi silver halide photosensitive material
JPS604184A (en) * 1983-06-23 1985-01-10 Chugai Pharmaceut Co Ltd Pyrazoloindazole derivative
JPH083564B2 (en) 1986-07-25 1996-01-17 三菱化学株式会社 Polarizing film
DE3842091A1 (en) * 1987-12-14 1989-07-13 Fuji Photo Film Co Ltd IMAGING PROCESS
EP0477436B1 (en) 1990-09-24 1995-12-13 Agfa-Gevaert N.V. Roomlight handleable UV sensitive direct positive silver halide photographic material
US5208248A (en) * 1991-01-11 1993-05-04 Merck Sharpe & Dohme, Ltd. Indazole-substituted five-membered heteroaromatic compounds
EP0494774A1 (en) 1991-01-11 1992-07-15 MERCK SHARP &amp; DOHME LTD. Indazole-substituted fivemembered heteroaromatic compounds
US5612360A (en) 1992-06-03 1997-03-18 Eli Lilly And Company Angiotensin II antagonists
AU5704594A (en) 1992-12-18 1994-07-19 Wellcome Foundation Limited, The Pyrimidine, pyridine, pteridinone and indazole derivatives as enzyme inhibitors
US5625031A (en) 1994-02-08 1997-04-29 Bristol-Myers Squibb Company Peptide inhibitors of the p33cdk2 and p34cdc2 cell cycle regulatory kinases and human papillomavirus E7 oncoprotein
US5631156A (en) 1994-06-21 1997-05-20 The University Of Michigan DNA encoding and 18 KD CDK6 inhibiting protein
DE69532817T2 (en) 1994-11-10 2005-01-13 Millenium Pharmaceuticals, Inc., Cambridge USE OF PYRAZOLE COMPOUNDS FOR THE TREATMENT OF GLOMERULONEPHRITIS, CANCER, ATHEROSCLEROSIS OR RESTENOSIS
ZA951822B (en) * 1994-12-23 1996-09-26 Glaxo Group Ltd Chemical compounds
KR19980701879A (en) 1995-02-02 1998-06-25 피터 기딩스 Indole derivatives as 5-hydroxytryptamine receptor antagonists
AU6526896A (en) 1995-07-22 1997-02-18 Rhone-Poulenc Rorer Limited Substituted aromatic compounds and their pharmaceutical use
US5733920A (en) 1995-10-31 1998-03-31 Mitotix, Inc. Inhibitors of cyclin dependent kinases
US5760028A (en) * 1995-12-22 1998-06-02 The Dupont Merck Pharmaceutical Company Integrin receptor antagonists
US6495526B2 (en) 1996-01-23 2002-12-17 Gpc Biotech, Inc. Inhibitors of cell-cycle progression and uses related thereto
WO1997034876A1 (en) 1996-03-15 1997-09-25 Zeneca Limited Cinnoline derivatives and use as medicine
GB9608435D0 (en) 1996-04-24 1996-06-26 Celltech Therapeutics Ltd Chemical compounds
US5849733A (en) 1996-05-10 1998-12-15 Bristol-Myers Squibb Co. 2-thio or 2-oxo flavopiridol analogs
US5821243A (en) 1996-07-22 1998-10-13 Viropharma Incorporated Compounds compositions and methods for treating influenza
GB9621757D0 (en) 1996-10-18 1996-12-11 Ciba Geigy Ag Phenyl-substituted bicyclic heterocyclyl derivatives and their use
WO1998033798A2 (en) 1997-02-05 1998-08-06 Warner Lambert Company Pyrido[2,3-d]pyrimidines and 4-amino-pyrimidines as inhibitors of cell proliferation
AU6230098A (en) 1997-02-27 1998-09-18 Tanabe Seiyaku Co., Ltd. Isoquinolinone derivatives, process for preparing the same, and their use as phosphodiesterase inhibitors
JP2001518094A (en) 1997-03-31 2001-10-09 デュポン ファーマシューティカルズ カンパニー Indazole-cyclic ureas useful as HIV protease inhibitors
CA2294244A1 (en) 1997-07-12 1999-01-21 Cancer Research Campaign Technology Limited Cyclin dependent kinase inhibiting purine derivatives
GB9716324D0 (en) 1997-08-01 1997-10-08 Mead Corp Packaging machine and method of carton set up
FR2767475A1 (en) 1997-08-25 1999-02-26 Oreal Oxidation hair dyes
FR2767827A1 (en) 1997-09-03 1999-02-26 Adir NOVEL INDOLE AND INDAZOLE DERIVATIVES, PROCESS FOR PREPARING THEM AND PHARMACEUTICAL COMPOSITIONS CONTAINING THE SAME
GB9718913D0 (en) * 1997-09-05 1997-11-12 Glaxo Group Ltd Substituted oxindole derivatives
DE19744026A1 (en) * 1997-10-06 1999-04-08 Hoechst Marion Roussel De Gmbh Pyrazole derivatives, their preparation and their use in medicinal products
AU9691198A (en) * 1997-10-06 1999-04-27 Basf Aktiengesellschaft Indeno{1,2-c}-, naphtho{1,2-c}- and benzo{6,7}cyclohepta{1,2-c}pyrazole derivatives
BR9815200A (en) 1997-10-27 2001-10-16 Agouron Pharma Compound, pharmaceutical composition and method of treating a disease or disorder mediated by the inhibition of cdk4 or a cdk4 / cyclin complex
IL135900A0 (en) 1997-11-04 2001-05-20 Pfizer Prod Inc Indazole bioisostere replacement of catechol in therapeuticaly active compounds
NZ503995A (en) 1997-11-04 2003-02-28 Pfizer Prod Inc Indazole compounds, and pharmaceutical compositions and uses thereof, based on indazole bioisostere replacement of catechol in PDE4 inhibitors
US6040321A (en) 1997-11-12 2000-03-21 Bristol-Myers Squibb Company Aminothiazole inhibitors of cyclin dependent kinases
HUP0100931A3 (en) 1998-02-26 2002-08-28 Aventis Pharma Inc 6,9-disubstituted 2-[trans-(-aminocyclohexyl)amino]purines and pharmaceutical compositions containing them
US6479487B1 (en) 1998-02-26 2002-11-12 Aventis Pharmaceuticals Inc. 6, 9-disubstituted 2-[trans-(4-aminocyclohexyl)amino] purines
EA003603B1 (en) 1998-02-27 2003-06-26 Пфайзер Продактс Инк. N-[(substituted five-membered di- or triaza diunsaturated ring)carbonyl] guanidine derivatives for the treatment of ischemia
JP4665239B2 (en) 1998-04-21 2011-04-06 アゲンニクス アーゲー 5-Aminoindeno [1,2-c] pyrazol-4-ones as anticancer and antiproliferative agents
YU54202A (en) * 2000-01-18 2006-01-16 Agouron Pharmaceuticals Inc. Indazole compounds,pharmaceutical compositions,and methods for mediating or inhibiting cell proliferation
US6897231B2 (en) * 2000-07-31 2005-05-24 Signal Pharmaceuticals, Inc. Indazole derivatives as JNK inhibitors and compositions and methods related thereto
EP1545515A1 (en) * 2002-08-12 2005-06-29 Sugen, Inc. 3-pyrrolyl-pyridopyrazoles and 3-pyrrolyl-indazoles as novel kinase inhibitors

Also Published As

Publication number Publication date
WO2001053268B1 (en) 2002-01-24
BG107011A (en) 2003-04-30
YU54202A (en) 2006-01-16
NZ518531A (en) 2004-09-24
DZ3301A1 (en) 2001-07-26
EE200200398A (en) 2003-10-15
NO20022117L (en) 2002-09-16
CA2388885A1 (en) 2001-07-26
CN1394205A (en) 2003-01-29
US6919461B2 (en) 2005-07-19
ZA200203040B (en) 2003-08-11
HUP0203965A2 (en) 2003-05-28
NO20022117D0 (en) 2002-05-03
CO5280070A1 (en) 2003-05-30
KR20020073505A (en) 2002-09-26
PL357590A1 (en) 2004-07-26
AP2002002564A0 (en) 2002-06-30
PA8509901A1 (en) 2002-08-26
CR6630A (en) 2004-01-29
SV2002000293A (en) 2002-12-02
EA200200768A1 (en) 2003-02-27
DOP2001000120A (en) 2002-04-15
UA75880C2 (en) 2006-06-15
MY136604A (en) 2008-10-31
JP2003520273A (en) 2003-07-02
PE20011334A1 (en) 2002-01-14
AR032438A1 (en) 2003-11-12
US7232912B2 (en) 2007-06-19
HUP0203965A3 (en) 2003-07-28
US20060111322A1 (en) 2006-05-25
US20030139463A1 (en) 2003-07-24
AU785013B2 (en) 2006-08-24
WO2001053268A3 (en) 2001-12-27
US6555539B2 (en) 2003-04-29
GT200100009A (en) 2002-03-14
OA12160A (en) 2006-05-08
GEP20043363B (en) 2004-05-10
US20020161022A1 (en) 2002-10-31
HN2001000007A (en) 2001-06-13
AP1609A (en) 2006-05-08
SK10052002A3 (en) 2003-03-04
WO2001053268A2 (en) 2001-07-26
AU2953901A (en) 2001-07-31
MA27589A1 (en) 2005-11-01
IS6474A (en) 2002-07-16
EP1250326A2 (en) 2002-10-23
BR0107783A (en) 2002-11-19
US20050239855A1 (en) 2005-10-27
HRP20020675A2 (en) 2004-12-31
IL150730A0 (en) 2003-02-12

Similar Documents

Publication Publication Date Title
MXPA02007058A (en) Indazole compounds, pharmaceutical compositions, and methods for mediating or inhibiting cell proliferation.
US6462069B2 (en) Compounds, pharmaceutical compositions, and methods for inhibiting protein kinases
JP4762543B2 (en) Indazole carboxamide derivatives, processes for their preparation and use as CDK1, CDK2 and CDK4 inhibitors
AU777701B2 (en) Indazole compounds and pharmaceutical compositions for inhibiting protein kinases, and methods for their use
CN102993084B (en) Alkynyl derivatives as modulators of metabotropic glutamate receptors
EP2468729A1 (en) Novel indazole derivative
KR20020015333A (en) Diaminothiazoles and their use for inhibiting protein kinases
WO2005094805A1 (en) Imine derivative and amide derivative
WO2005090328A1 (en) Heterocyclic compound and use thereof
CN105073742A (en) Flap modulators
US6566363B2 (en) Pyrazole-thiazole compounds, pharmaceutical compositions containing them
JPH10291988A (en) Heterocyclic compound
EP4023640A1 (en) Phenylimidazole compound

Legal Events

Date Code Title Description
FG Grant or registration